Epigenetic Regulation of Aging and its Rejuvenation

Yongpan An , Qian Wang , Ke Gao , Chi Zhang , Yanan Ouyang , Ruixiao Li , Zhou Ma , Tong Wu , Lifan Zhou , Zhengwei Xie , Rui Zhang , Guojun Wu

MedComm ›› 2025, Vol. 6 ›› Issue (9) : e70369

PDF
MedComm ›› 2025, Vol. 6 ›› Issue (9) : e70369 DOI: 10.1002/mco2.70369
REVIEW

Epigenetic Regulation of Aging and its Rejuvenation

Author information +
History +
PDF

Abstract

Aging increases the global burden of disease, yet its molecular basis remains incompletely understood. Recent studies indicate that reversible epigenetic drift—spanning DNA methylation clocks, histone codes, three-dimensional chromatin, and noncoding RNA networks—constitutes a central regulator of organismal decline and age-related diseases. How these epigenetic layers interact across different tissues—and how best to translate them into therapeutic strategies—are still open questions. This review outlines the specific mechanisms by which epigenetic changes influence aging, highlighting their impact on genomic instability, stem-cell exhaustion, and mitochondrial dysfunction. We critically evaluate emerging rejuvenation strategies—partial OSKM reprogramming, CRISPR–dCas9 epigenome editing, NAD⁺/sirtuin boosters, HDAC inhibitors, microbiota transfer, and precision lifestyle interventions—detailing their efficacy in resetting epigenetic age and restoring tissue homeostasis. Integrating single-cell multiomics and second-generation epigenetic clocks, we propose a roadmap for translating these insights into safe, personalized antiaging medicine.

Keywords

aging / epigenetic rejuvenation / epigenetic clocks / epigenetic mechanisms / emerging rejuvenation strategies

Cite this article

Download citation ▾
Yongpan An, Qian Wang, Ke Gao, Chi Zhang, Yanan Ouyang, Ruixiao Li, Zhou Ma, Tong Wu, Lifan Zhou, Zhengwei Xie, Rui Zhang, Guojun Wu. Epigenetic Regulation of Aging and its Rejuvenation. MedComm, 2025, 6(9): e70369 DOI:10.1002/mco2.70369

登录浏览全文

4963

注册一个新账户 忘记密码

References

[1]

Y. Cai, W. Song, J. Li, et al., “The Landscape of Aging,” Sci China Life Sci 65 (2022): 2354-2454.

[2]

A. Scott, M. Ellison, and D. Sinclair, “The Economic Value of Targeting Aging,” Nat Aging 1 (2021): 616-623.

[3]

C. Lopez-Otin, M. Blasco, L. Partridge, M. Serrano, and G. Kroemer, “Hallmarks of Aging: An Expanding Universe,” Cell 186 (2023): 243-278.

[4]

J. Yang, M. Hayano, P. Griffin, et al., “Loss of Epigenetic Information as a Cause of Mammalian Aging,” Cell 186 (2023): 305-326.

[5]

T. Eisenberg, H. Knauer, A. Schauer, et al., “Induction of Autophagy by Spermidine Promotes Longevity,” Nature Cell Biology 11 (2009): 1305-1314.

[6]

W. Zhang, J. Qu, G. Liu, and J. Belmonte, “The Ageing Epigenome and Its Rejuvenation,” Nature Reviews Molecular Cell Biology 21 (2020): 137-150.

[7]

A. Parkhitko, E. Filine, and M. Tatar, “Combinatorial Interventions in Aging,” Nat Aging 3 (2023): 1187-1200.

[8]

S. Fadul, A. Arshad, and R. Mehmood, “CRISPR-based Epigenome Editing: Mechanisms and Applications,” Epigenomics 15 (2023): 1137-1155.

[9]

K. Seale, S. Horvath, A. Teschendorff, N. Eynon, and S. Voisin, “Making Sense of the Ageing Methylome,” Nature Reviews Genetics 23 (2022): 585-605.

[10]

S. Smith, B. Kaplan, L. Sowers, and E. Newman, “Mechanism of human Methyl-directed DNA Methyltransferase and the Fidelity of Cytosine Methylation,” PNAS 89 (1992): 4744-4748.

[11]

X. Ming, Z. Zhang, Z. Zou, et al., “Kinetics and Mechanisms of Mitotic Inheritance of DNA Methylation and Their Roles in Aging-associated Methylome Deterioration,” Cell Research 30 (2020): 980-996.

[12]

S. Johnstone, V. Gladyshev, M. Aryee, and B. Bernstein, “Epigenetic Clocks, Aging, and Cancer,” Science 378 (2022): 1276-1277.

[13]

L. Chouliaras, D. van den Hove, G. Kenis, et al., “Prevention of Age-related Changes in Hippocampal Levels of 5-methylcytidine by Caloric Restriction,” Neurobiology of Aging 33 (2012): 1672-1681.

[14]

A. Unnikrishnan, W. Freeman, J. Jackson, J. Wren, H. Porter, and A. Richardson, “The Role of DNA Methylation in Epigenetics of Aging,” Pharmacology & Therapeutics 195 (2019): 172-185.

[15]

A. Bick, J. Pirruccello, G. Griffin, et al., “Genetic Interleukin 6 Signaling Deficiency Attenuates Cardiovascular Risk in Clonal Hematopoiesis,” Circulation 141 (2020): 124-131.

[16]

C. Chen and H. Huang, “Methyltransferase 3-mediated m6A Modification of Switch/Sucrose Non-fermenting-related Matrix-associated Actin-dependent Regulator of Chromatin Subfamily a Member 5 Promotes Mycobacterium Tuberculosis-infected Macrophage M1 Polarization and Inflammation,” Cytojournal 22 (2025): 38.

[17]

Z. Zhang, R. Lu, P. Wang, et al., “Structural Basis for DNMT3A-mediated De Novo DNA Methylation,” Nature 554 (2018): 387-391.

[18]

M. Yagi, M. Kabata, A. Tanaka, et al., “Identification of Distinct Loci for De Novo DNA Methylation by DNMT3A and DNMT3B During Mammalian Development,” Nature Communications 11 (2020): 3199.

[19]

A. Lu, Z. Fei, A. Haghani, et al., “Universal DNA Methylation Age Across Mammalian Tissues,” Nat Aging 3 (2023): 1144-1166.

[20]

B. Bhargavan, B. Chhunchha, E. Kubo, and D. Singh, “DNA Methylation as an Epigenetic Mechanism in the Regulation of LEDGF Expression and Biological Response in Aging and Oxidative Stress,” Cell Death Discov 10 (2024): 296.

[21]

A. So, J. Jung, S. Lee, H. Kim, and K. Kang, “DNA Methyltransferase Controls Stem Cell Aging by Regulating BMI1 and EZH2 Through microRNAs,” PLoS ONE 6 (2011): e19503.

[22]

K. Wang, H. Liu, Q. Hu, et al., “Epigenetic Regulation of Aging: Implications for Interventions of Aging and Diseases,” Signal Transduct Target Ther 7 (2022): 374.

[23]

I. Shchukina, J. Bagaitkar, O. Shpynov, et al., “Enhanced Epigenetic Profiling of Classical human Monocytes Reveals a Specific Signature of Healthy Aging in the DNA Methylome,” Nat Aging 1 (2021): 124-141.

[24]

S. Glaser, R. Wagener, H. Kretzmer, et al., “Subtyping Burkitt Lymphoma by DNA Methylation,” Genes, chromosomes & cancer 64 (2025): e70042.

[25]

N. Song, E. Ji, J. Yu, et al., “Spermidine Enhances Mitochondrial Function and Mitigates Aortic Valve Calcification: Implications for DNA Methyltransferase-1 Activity,” JACC Basic Transl Sci 10 (2025): 345-366.

[26]

J. Xiong, F. Ma, N. Ding, et al., “p Alleviates Homocysteine-mediated Atherosclerosis by Targeting IL-31 Through Its Epigenetics Modifications,” Aging Cell 20 (2021): e13485.

[27]

L. Wang, L. Lankhorst, and R. Bernards, “Exploiting Senescence for the Treatment of Cancer,” Nature Reviews Cancer 22 (2022): 340-355.

[28]

K. Yan, Q. Ji, D. Zhao, et al., “SGF29 nuclear Condensates Reinforce Cellular Aging,” Cell Discovery 9 (2023): 110.

[29]

B. Zhang, Q. Long, S. Wu, et al., “KDM4 Orchestrates Epigenomic Remodeling of Senescent Cells and Potentiates the Senescence-Associated Secretory Phenotype,” Nat Aging 1 (2021): 454-472.

[30]

W. Wang, Y. Zheng, S. Sun, et al., “A Genome-wide CRISPR-based Screen Identifies KAT7 as a Driver of Cellular Senescence,” Science Translational Medicine 13 (2021): eabd2655.

[31]

F. Della Valle, P. Reddy, M. Yamamoto, et al., “LINE-1 RNA Causes Heterochromatin Erosion and Is a Target for Amelioration of Senescent Phenotypes in Progeroid Syndromes,” Science Translational Medicine 14 (2022): eabl6057.

[32]

Z. Liu, Q. Ji, J. Ren, et al., “Large-scale Chromatin Reorganization Reactivates Placenta-specific Genes That Drive Cellular Aging,” Developmental Cell 57 (2022): 1347-1368. e1312.

[33]

V. Gorgoulis, P. Adams, A. Alimonti, et al., “Cellular Senescence: Defining a Path Forward,” Cell 179 (2019): 813-827.

[34]

A. Trapp, C. Kerepesi, and V. Gladyshev, “Profiling Epigenetic Age in Single Cells,” Nat Aging 1 (2021): 1189-1201.

[35]

K. Rudolph, “DNA-methylation Aging at Single-cell Level,” Nat Aging 1 (2021): 1086-1087.

[36]

K. Margiotti, F. Monaco, M. Fabiani, A. Mesoraca, and C. Giorlandino, “Epigenetic Clocks: In Aging-Related and Complex Diseases,” Cytogenetic and Genome Research 163 (2023): 247-256.

[37]

C. Vatier and S. Christin-Maitre, “Epigenetic/Circadian Clocks and PCOS,” Human Reproduction 39 (2024): 1167-1175.

[38]

Z. Wu, J. Qu, W. Zhang, and G. Liu, “Stress, Epigenetics, and Aging: Unraveling the Intricate Crosstalk,” Molecular Cell 84 (2024): 34-54.

[39]

R. Duan, Q. Fu, Y. Sun, and Q. Li, “Epigenetic Clock: A Promising Biomarker and Practical Tool in Aging,” Ageing Research Reviews 81 (2022): 101743.

[40]

S. Horvath, “DNA Methylation Age of human Tissues and Cell Types,” Genome biology 14 (2013): R115.

[41]

G. Hannum, J. Guinney, L. Zhao, et al., “Genome-wide Methylation Profiles Reveal Quantitative Views of human Aging Rates,” Molecular Cell 49 (2013): 359-367.

[42]

M. Levine, A. Lu, A. Quach, et al., “An Epigenetic Biomarker of Aging for Lifespan and Healthspan,” Aging (Albany NY) 10 (2018): 573-591.

[43]

A. Lu, A. Quach, J. Wilson, et al., “DNA Methylation GrimAge Strongly Predicts Lifespan and Healthspan,” Aging (Albany NY) 11 (2019): 303-327.

[44]

D. Belsky, A. Caspi, D. Corcoran, et al., “DunedinPACE, a DNA Methylation Biomarker of the Pace of Aging,” Elife 11 (2022): e73420.

[45]

T. Yang, Y. Xiao, Y. Cheng, et al., “Epigenetic Clocks in Neurodegenerative Diseases: A Systematic Review,” Journal of Neurology, Neurosurgery, and Psychiatry 94 (2023): 1064-1070.

[46]

K. Murach, A. Dimet-Wiley, Y. Wen, et al., “Late-life Exercise Mitigates Skeletal Muscle Epigenetic Aging,” Aging Cell 21 (2022): e13527.

[47]

W. Zhao, S. Yu, Y. Xu, et al., “Sleep Traits Causally Affect Epigenetic Age Acceleration: A Mendelian Randomization Study,” Scientific Reports 15 (2025): 7439.

[48]

T. Perlstein, J. Jung, A. Wagner, et al., “Alcohol and Aging: Next-generation Epigenetic Clocks Predict Biological Age Acceleration in Individuals With Alcohol Use Disorder,” Alcohol Clin Exp Res (Hoboken) 49 (2025): 829-842.

[49]

H. Sung and W. Lin, “Causal Effects of Cardiovascular Health on Five Epigenetic Clocks,” Clin Epigenetics 16 (2024): 134.

[50]

Y. Lo and W. Lin, “Cardiovascular Health and Four Epigenetic Clocks,” Clin Epigenetics 14 (2022): 73.

[51]

A. Bienkowska, G. Raddatz, J. Sohle, et al., “Development of an Epigenetic Clock to Predict Visual Age Progression of human Skin,” Front Aging 4 (2023): 1258183.

[52]

D. Kim, J. Kang, J. Kim, et al., “Assessing the Utility of Epigenetic Clocks for Health Prediction in South Korean,” Front Aging 5 (2024): 1493406.

[53]

J. Peng, C. Yuan, X. Hua, and Z. Zhang, “Molecular Mechanism of Histone Variant H2A.B on Stability and Assembly of Nucleosome and Chromatin Structures,” Epigenetics & chromatin 13 (2020): 28.

[54]

B. Schumacher, J. Pothof, J. Vijg, and J. Hoeijmakers, “The central Role of DNA Damage in the Ageing Process,” Nature 592 (2021): 695-703.

[55]

P. Mustafi, M. Hu, S. Kumari, C. Das, G. Li, and T. Kundu, “Phosphorylation-dependent Association of human Chromatin Protein PC4 to Linker Histone H1 Regulates Genome Organization and Transcription,” Nucleic Acids Res. 50 (2022): 6116-6136.

[56]

Y. Lu, X. Tian, and D. Sinclair, “The Information Theory of Aging,” Nat Aging 3 (2023): 1486-1499.

[57]

G. Millan-Zambrano, A. Burton, A. Bannister, and R. Schneider, “Histone Post-translational Modifications—cause and Consequence of Genome Function,” Nature Reviews Genetics 23 (2022): 563-580.

[58]

A. Patel, Y. He, and I. Radhakrishnan, “Histone Acetylation and Deacetylation—Mechanistic Insights From Structural Biology,” Gene 890 (2024): 147798.

[59]

R. Liu, J. Wu, H. Guo, et al., “Post-translational Modifications of Histones: Mechanisms, Biological Functions, and Therapeutic Targets,” MedComm 2023 (2020): e292.

[60]

K. Bonitto, K. Sarathy, K. Atai, M. Mitra, and H. Coller, “Is There a Histone Code for Cellular Quiescence?” Frontiers in Cell and Developmental Biology 9 (2021): 739780.

[61]

N. Fuggle, F. Laskou, N. Harvey, and E. Dennison, “A Review of Epigenetics and Its Association With Ageing of Muscle and Bone,” Maturitas 165 (2022): 12-17.

[62]

S. Mishra, M. Raval, A. Kachhawaha, B. Tiwari, and A. Tiwari, “Aging: Epigenetic Modifications,” Progress in Molecular Biology and Translational Science 197 (2023): 171-209.

[63]

C. Soto-Palma, L. Niedernhofer, C. Faulk, and X. Dong, “Epigenetics, DNA Damage, and Aging,” Journal of Clinical Investigation 132 (2022): e158446.

[64]

Y. Zhang, Z. Sun, J. Jia, et al., “Overview of Histone Modification,” Advances in Experimental Medicine and Biology 1283 (2021): 1-16.

[65]

A. Zhao, W. Xu, R. Han, et al., “Role of Histone Modifications in Neurogenesis and Neurodegenerative Disease Development,” Ageing Research Reviews 98 (2024): 102324.

[66]

K. Weaver, R. Holt, E. Henry, Y. Lyu, and S. Pletcher, “Effects of Hunger on Neuronal Histone Modifications Slow Aging in Drosophila,” Science 380 (2023): 625-632.

[67]

Y. Sun, H. Zhang, T. Qiu, L. Liao, and X. Su, “Epigenetic Regulation of Mesenchymal Stem Cell Aging Through Histone Modifications,” Genes Dis 10 (2023): 2443-2456.

[68]

A. la Torre, F. Lo Vecchio, and A. Greco, “Epigenetic Mechanisms of Aging and Aging-Associated Diseases,” Cells 12 (2023): 1163.

[69]

M. Al Ojaimi, B. Banimortada, A. Othman, K. Riedhammer, M. Almannai, and A. El-Hattab, “Disorders of Histone Methylation: Molecular Basis and Clinical Syndromes,” Clinical Genetics 102 (2022): 169-181.

[70]

D. Sun, M. Luo, M. Jeong, et al., “Epigenomic Profiling of Young and Aged HSCs Reveals Concerted Changes During Aging That Reinforce Self-renewal,” Cell Stem Cell 14 (2014): 673-688.

[71]

N. Itokawa, M. Oshima, S. Koide, et al., “Epigenetic Traits Inscribed in Chromatin Accessibility in Aged Hematopoietic Stem Cells,” Nature Communications 13 (2022): 2691.

[72]

B. Benayoun, E. Pollina, P. Singh, et al., “Remodeling of Epigenome and Transcriptome Landscapes With Aging in Mice Reveals Widespread Induction of Inflammatory Responses,” Genome Research 29 (2019): 697-709.

[73]

L. Liu, T. Cheung, G. Charville, et al., “Chromatin Modifications as Determinants of Muscle Stem Cell Quiescence and Chronological Aging,” Cell reports 4 (2013): 189-204.

[74]

P. Sen, P. Shah, R. Nativio, and S. Berger, “Epigenetic Mechanisms of Longevity and Aging,” Cell 166 (2016): 822-839.

[75]

P. Cheung, F. Vallania, H. Warsinske, et al., “Single-Cell Chromatin Modification Profiling Reveals Increased Epigenetic Variations With Aging,” Cell 173 (2018): 1385-1397. e1314.

[76]

J. Li, J. Li, and E. Guo, “Quercetin Suppresses Glioma Stem Cells via Activating p16-INK4 Gene Expression Through Epigenetic Regulation,” Anti-cancer agents in medicinal chemistry 25, no. 14 (2025): 1041-1048, https://doi.org/10.2174/0118715206332048241126095207.

[77]

Y. Li, G. Fang, W. Cao, et al., “Ezh2 Inhibits Replicative Senescence of Atrial Fibroblasts through Promotion of H3K27me3 in the Promoter Regions of CDKN2a and Timp4 Genes,” J Inflamm Res 15 (2022): 4693-4708.

[78]

T. Yasuda, M. Koiwa, A. Yonemura, et al., “Inflammation-driven Senescence-associated Secretory Phenotype in Cancer-associated Fibroblasts Enhances Peritoneal Dissemination,” Cell reports 34 (2021): 108779.

[79]

S. Sugita, T. Aoyama, M. Emori, et al., “Assessment of H3K27me3 Immunohistochemistry and Combination of NF1 and p16 Deletions by Fluorescence in Situ Hybridization in the Differential Diagnosis of Malignant Peripheral Nerve Sheath Tumor and Its Histological Mimics,” Diagn Pathol 16 (2021): 79.

[80]

H. Zhang, J. Li, Y. Yu, et al., “Nuclear Lamina Erosion-induced Resurrection of Endogenous Retroviruses Underlies Neuronal Aging,” Cell reports 42 (2023): 113396.

[81]

F. Xiao, H. Wang, X. Chen, et al., “Hypermethylation in H3K9me3 Regions Characterizes the Centenarian Methylomes in Healthy Aging,” National Science Review 10 (2023): nwad067.

[82]

Q. Wan, X. Meng, C. Wang, et al., “Histone H3K4me3 Modification Is a Transgenerational Epigenetic Signal for Lipid Metabolism in Caenorhabditis elegans,” Nature Communications 13 (2022): 768.

[83]

Y. Guo, S. Zhao, and G. Wang, “Polycomb Gene Silencing Mechanisms: PRC2 Chromatin Targeting, H3K27me3 'Readout', and Phase Separation-Based Compaction,” Trends in Genetics 37 (2021): 547-565.

[84]

S. Ro, “Improving Gastric Motility in Aging through EZH2 Inhibition and Preservation of Interstitial Cells of Cajal,” Cell Mol Gastroenterol Hepatol 18 (2024): 101382.

[85]

R. Kumbhar, A. Sanchez, J. Perren, et al., “Poly(ADP-ribose) Binding and macroH2A Mediate Recruitment and Functions of KDM5A at DNA Lesions,” Journal of Cell Biology 220 (2021): e202006149.

[86]

A. Godbole, S. Gopalan, T. Nguyen, et al., “S-adenosylmethionine Synthases Specify Distinct H3K4me3 Populations and Gene Expression Patterns During Heat Stress,” Elife 12 (2023): e79511.

[87]

Y. Xiao, F. Liu, Q. Kong, et al., “Metformin Induces S-adenosylmethionine Restriction to Extend the Caenorhabditis elegans Healthspan Through H3K4me3 Modifiers,” Aging Cell 21 (2022): e13567.

[88]

Y. Zhang, R. Ma, Q. Deng, et al., “S-adenosylmethionine Improves Cognitive Impairment in D-galactose-induced Brain Aging by Inhibiting Oxidative Stress and Neuroinflammation,” Journal of Chemical Neuroanatomy 128 (2023): 102232.

[89]

Z. Gu, Y. Wang, Z. Fang, et al., “Plasma Metabolomics Identifies S-adenosylmethionine as a Biomarker and Potential Therapeutic Target for Vascular Aging in Older Adult Males,” Journal of Pharmaceutical and Biomedical Analysis 243 (2024): 116097.

[90]

S. Okstad, I. Mair, J. Sundsfjord, T. Eide, and I. Nordrum, “[Ectopic thyroid tissue in the head and neck],” Tidsskrift for Den Norske Laegeforening 107 (1987): 249-250.

[91]

T. Chen, F. Wang, P. Lee, A. Hsu, and T. Ching, “Mitochondrial S-adenosylmethionine Deficiency Induces Mitochondrial Unfolded Protein Response and Extends Lifespan in Caenorhabditis elegans,” Aging Cell 23 (2024): e14103.

[92]

A. Bellver-Sanchis, Q. Geng, G. Navarro, et al., “G9a Inhibition Promotes Neuroprotection Through GMFB Regulation in Alzheimer's Disease,” Aging Dis 15 (2024): 311-337.

[93]

B. Signal, A. Phipps, K. Giles, et al., “Ageing-Related Changes to H3K4me3, H3K27ac, and H3K27me3 in Purified Mouse Neurons,” Cells 13 (2024): 1393.

[94]

Z. Yang, D. Yu, F. Gao, et al., “The Histone Lysine Demethylase KDM7A Contributes to Reward Memory via Fscn1-Induced Synaptic Plasticity in the Medial Prefrontal Cortex,” Adv Sci (Weinh) 12 (2025): e2405352.

[95]

L. Xiao, J. Qiao, Y. Huang, et al., “RASGRP1 targeted by H3K27me3 Regulates Myoblast Proliferation and Differentiation in Mice and Pigs,” Acta Biochim Biophys Sin (Shanghai) 56 (2024): 452-461.

[96]

J. Shimizu and F. Kawano, “DNA Hypermethylation Preceded by H3K27 Trimethylation Is Linked to Downregulation of Gene Expression in Disuse Muscle Atrophy in Male Mice,” Physiological Reports 13 (2025): e70317.

[97]

M. Izadi, N. Sadri, A. Abdi, S. Serajian, D. Jalalei, and S. Tahmasebi, “Epigenetic Biomarkers in Aging and Longevity: Current and Future Application,” Life Sciences 351 (2024): 122842.

[98]

Z. Sun, Y. Zhang, J. Jia, et al., “H3K36me3, message From Chromatin to DNA Damage Repair,” Cell Biosci 10 (2020): 9.

[99]

L. Su, H. Li, C. Huang, et al., “Muscle-Specific Histone H3K36 Dimethyltransferase SET-18 Shortens Lifespan of Caenorhabditis elegans by Repressing Daf-16a Expression,” Cell reports 22 (2018): 2716-2729.

[100]

L. Ni, B. Lin, Y. Zhang, et al., “Histone Modification Landscape and the Key Significance of H3K27me3 in Myocardial Ischaemia/Reperfusion Injury,” Sci China Life Sci 66 (2023): 1264-1279.

[101]

M. Bouwman, D. de Bakker, H. Honkoop, et al., “Cross-species Comparison Reveals That Hmga1 Reduces H3K27me3 Levels to Promote Cardiomyocyte Proliferation and Cardiac Regeneration,” Nat Cardiovasc Res 4 (2025): 64-82.

[102]

J. Cordero, A. Elsherbiny, Y. Wang, et al., “Leveraging Chromatin state Transitions for the Identification of Regulatory Networks Orchestrating Heart Regeneration,” Nucleic Acids Res. 52 (2024): 4215-4233.

[103]

C. Bonfiglio, M. Lacy, V. Triantafyllidou, et al., “Ezh2 Shapes T Cell Plasticity to Drive Atherosclerosis,” Circulation 151, no. 19 (2025): 1391-1408, https://doi.org/10.1161/CIRCULATIONAHA.124.072384.

[104]

P. Lu, J. Xu, X. Shen, et al., “Spatiotemporal Role of SETD2-H3K36me3 in Murine Pancreatic Organogenesis,” Cell reports 43 (2024): 113703.

[105]

S. Guo, J. Fang, W. Xu, et al., “Interplay Between H3K36me3, Methyltransferase SETD2, and Mismatch Recognition Protein MutSalpha Facilitates Processing of Oxidative DNA Damage in human Cells,” Journal of Biological Chemistry 298 (2022): 102102.

[106]

A. Guerrero, A. Innes, P. Roux, et al., “3-deazaadenosine (3DA) Alleviates Senescence to Promote Cellular Fitness and Cell Therapy Efficiency in Mice,” Nat Aging 2 (2022): 851-866.

[107]

X. Yi, Q. Zhu, X. Wu, T. Tan, and X. Jiang, “Histone Methylation and Oxidative Stress in Cardiovascular Diseases,” Oxid Med Cell Longev 2022 (2022): 6023710.

[108]

M. Huang, Q. Wu, and Z. Jiang, “Epigenetic Alterations Under Oxidative Stress in Stem Cells,” Oxid Med Cell Longev 2022 (2022): 6439097.

[109]

X. Zheng and A. Sawalha, “The Role of Oxidative Stress in Epigenetic Changes Underlying Autoimmunity,” Antioxid Redox Signaling 36 (2022): 423-440.

[110]

C. Wang, B. Hu, Y. Yang, et al., “Structural Simulation and Selective Inhibitor Discovery Study for Histone Demethylases KDM4E/6B From a Computational Perspective,” Computational Biology and Chemistry 110 (2024): 108072.

[111]

Z. Wu, W. Zhang, J. Qu, and G. Liu, “Emerging Epigenetic Insights Into Aging Mechanisms and Interventions,” Trends in Pharmacological Sciences 45 (2024): 157-172.

[112]

J. Xu, C. Li, and X. Kang, “The Epigenetic Regulatory Effect of Histone Acetylation and Deacetylation on Skeletal Muscle Metabolism-a Review,” Front Physiol 14 (2023): 1267456.

[113]

J. Ouyang, D. Wu, Y. Gan, Y. Tang, H. Wang, and J. Huang, “Unraveling the Metabolic‒Epigenetic Nexus: A New Frontier in Cardiovascular Disease Treatment,” Cell death & disease 16 (2025): 183.

[114]

J. Zhang, F. Chen, Y. Tian, et al., “PARylated PDHE1alpha Generates Acetyl-CoA for Local Chromatin Acetylation and DNA Damage Repair,” Nature structural & molecular biology 30 (2023): 1719-1734.

[115]

P. Bradshaw, “Acetyl-CoA Metabolism and Histone Acetylation in the Regulation of Aging and Lifespan,” Antioxidants (Basel) 10 (2021): 572.

[116]

S. Serio, C. Pagiatakis, E. Musolino, et al., “Cardiac Aging Is Promoted by Pseudohypoxia Increasing p300-Induced Glycolysis,” Circulation Research 133 (2023): 687-703.

[117]

L. Qiu, X. Liu, H. Xia, and C. Xu, “Downregulation of P300/CBP-Associated Factor Protects From Vascular Aging via Nrf2 Signal Pathway Activation,” International Journal of Molecular Sciences 23 (2022): 12574.

[118]

A. Price, M. Manjegowda, J. Kain, S. Anandh, and I. Bochkis, “Hdac3, Setdb1, and Kap1 Mark H3K9me3/H3K14ac Bivalent Regions in Young and Aged Liver,” Aging Cell 19 (2020): e13092.

[119]

T. Pandita, C. Hunt, V. Singh, et al., “Role of the Histone Acetyl Transferase MOF and the Histone Deacetylase Sirtuins in Regulation of H4K16ac during DNA Damage Repair and Metabolic Programming: Implications in Cancer and Aging,” Sub-Cellular Biochemistry 100 (2022): 115-141.

[120]

Y. You and W. Liang, “SIRT1 and SIRT6: The Role in Aging-related Diseases,” Biochim Biophys Acta Mol Basis Dis 1869 (2023): 166815.

[121]

J. Chen, A. Wang, and Q. Chen, “SirT3 and p53 Deacetylation in Aging and Cancer,” Journal of Cellular Physiology 232 (2017): 2308-2311.

[122]

Z. Ji, G. Liu, and J. Qu, “Mitochondrial Sirtuins, Metabolism, and Aging,” J Genet Genomics 49 (2022): 287-298.

[123]

Y. Ye, K. Yang, H. Liu, et al., “SIRT2 counteracts Primate Cardiac Aging via Deacetylation of STAT3 That Silences CDKN2B,” Nat Aging 3 (2023): 1269-1287.

[124]

Y. Ding, T. Wang, S. Lv, et al., “SIRT6 is an Epigenetic Repressor of Thoracic Aortic Aneurysms via Inhibiting Inflammation and Senescence,” Signal Transduct Target Ther 8 (2023): 255.

[125]

Z. Guo, P. Li, J. Ge, and H. Li, “SIRT6 in Aging, Metabolism, Inflammation and Cardiovascular Diseases,” Aging Dis 13 (2022): 1787-1822.

[126]

O. Lozoya, T. Wang, D. Grenet, et al., “Mitochondrial Acetyl-CoA Reversibly Regulates Locus-specific Histone Acetylation and Gene Expression,” Life Sci Alliance 2 (2019): e201800228.

[127]

W. Li, Q. Long, H. Wu, et al., “Nuclear Localization of Mitochondrial TCA Cycle Enzymes Modulates Pluripotency via Histone Acetylation,” Nature Communications 13 (2022): 7414.

[128]

C. Chen, M. Zhou, Y. Ge, and X. Wang, “SIRT1 and Aging Related Signaling Pathways,” Mechanisms of Ageing and Development 187 (2020): 111215.

[129]

A. Covarrubias, R. Perrone, A. Grozio, and E. Verdin, “NAD(+) Metabolism and Its Roles in Cellular Processes During Ageing,” Nature Reviews Molecular Cell Biology 22 (2021): 119-141.

[130]

K. Joshi, S. Liu, S. Breslin, and J. Zhang, “Mechanisms That Regulate the Activities of TET Proteins,” Cellular and Molecular Life Sciences 79 (2022): 363.

[131]

Y. Feng, L. Cai, W. Hong, et al., “Rewiring of 3D Chromatin Topology Orchestrates Transcriptional Reprogramming and the Development of Human Dilated Cardiomyopathy,” Circulation 145 (2022): 1663-1683.

[132]

R. Li and X. Lin, “Connected Chromatin Amplifies Acetylation-Modulated Nucleosome Interactions,” Biochemistry 64 (2025): 1222-1232.

[133]

C. Dube, F. Jahan, and C. Lim, “Key Changes in Chromatin Mark Mammalian Epidermal Differentiation and Ageing,” Epigenetics 17 (2022): 444-459.

[134]

M. Shvedunova and A. Akhtar, “Modulation of Cellular Processes by Histone and Non-histone Protein Acetylation,” Nature Reviews Molecular Cell Biology 23 (2022): 329-349.

[135]

Y. Lin, A. Lin, L. Cai, et al., “ACSS2-dependent Histone Acetylation Improves Cognition in Mouse Model of Alzheimer's disease,” Mol Neurodegener 18 (2023): 47.

[136]

D. Dai, J. Xie, Y. Zheng, F. Chen, B. Zhao, and L. Miao, “H3K27 acetylation-induced FSTL1 Upregulation by P300/RUNX1 co-activation Exacerbated Autophagy-mediated Neuronal Damage and NF-kappaB-stimulated Inflammation in Alzheimer's disease,” Cytotechnology 75 (2023): 449-460.

[137]

S. Marzi, S. Leung, T. Ribarska, et al., “A Histone Acetylome-wide Association Study of Alzheimer's Disease Identifies Disease-associated H3K27ac Differences in the Entorhinal Cortex,” Nature Neuroscience 21 (2018): 1618-1627.

[138]

A. Greissel, M. Culmes, R. Burgkart, et al., “Histone Acetylation and Methylation Significantly Change With Severity of Atherosclerosis in human Carotid Plaques,” Cardiovascular Pathology 25 (2016): 79-86.

[139]

H. Kai, Q. Wu, R. Yin, et al., “LncRNA NORAD Promotes Vascular Endothelial Cell Injury and Atherosclerosis through Suppressing VEGF Gene Transcription via Enhancing H3K9 Deacetylation by Recruiting HDAC6,” Frontiers in Cell and Developmental Biology 9 (2021): 701628.

[140]

Y. Zhu, L. Shang, Y. Tang, et al., “Genome-Wide Profiling of H3K27ac Identifies TDO2 as a Pivotal Therapeutic Target in Metabolic Associated Steatohepatitis Liver Disease,” Adv Sci (Weinh) 11 (2024): e2404224.

[141]

Q. Song, X. Zhou, K. Xu, S. Liu, X. Zhu, and J. Yang, “The Safety and Antiaging Effects of Nicotinamide Mononucleotide in Human Clinical Trials: An Update,” Advances in nutrition 14 (2023): 1416-1435.

[142]

L. Guarente, D. Sinclair, and G. Kroemer, “Human Trials Exploring Anti-aging Medicines,” Cell metabolism 36 (2024): 354-376.

[143]

K. Freeberg, C. Udovich, C. Martens, D. Seals, and D. Craighead, “Dietary Supplementation with NAD+-Boosting Compounds in Humans: Current Knowledge and Future Directions,” Journals of Gerontology. Series A, Biological Sciences and Medical Sciences 78 (2023): 2435-2448.

[144]

Y. Qiu, Q. Xu, P. Xie, et al., “Epigenetic Modifications and Emerging Therapeutic Targets in Cardiovascular Aging and Diseases,” Pharmacological Research 211 (2025): 107546.

[145]

N. Gasek, G. Kuchel, J. Kirkland, and M. Xu, “Strategies for Targeting Senescent Cells in Human Disease,” Nat Aging 1 (2021): 870-879.

[146]

A. Sharma, S. Chabloz, R. Lapides, E. Roider, and C. Ewald, “Potential Synergistic Supplementation of NAD+ Promoting Compounds as a Strategy for Increasing Healthspan,” Nutrients 15 (2023): 445.

[147]

D. Wilson, M. Cookson, L. Van Den Bosch, H. Zetterberg, D. Holtzman, and , I. Dewachter, “Hallmarks of Neurodegenerative Diseases,” Cell 186 (2023): 693-714.

[148]

A. Sharma, K. Singh, and A. Almasan, “Histone H2AX Phosphorylation: A Marker for DNA Damage,” Methods in Molecular Biology 920 (2012): 613-626.

[149]

T. Oizumi, T. Suzuki, J. Kobayashi, and A. Nakamura, “Senescence-Associated Heterochromatin Foci Suppress Gamma-H2AX Focus Formation Induced by Radiation Exposure,” International Journal of Molecular Sciences 25 (2024): 3355.

[150]

T. Liu, J. Lin, C. Chen, et al., “MicroRNA-146b-5p Overexpression Attenuates Premature Ovarian Failure in Mice by Inhibiting the Dab2ip/Ask1/p38-Mapk Pathway and gammaH2A.X Phosphorylation,” Cell Proliferation 54 (2021): e12954.

[151]

S. Maity, K. Farrell, S. Navabpour, S. Narayanan, and T. Jarome, “Epigenetic Mechanisms in Memory and Cognitive Decline Associated With Aging and Alzheimer's Disease,” International Journal of Molecular Sciences 22 (2021): 12280.

[152]

A. Kinner, W. Wu, C. Staudt, and G. Iliakis, “Gamma-H2AX in Recognition and Signaling of DNA Double-strand Breaks in the Context of Chromatin,” Nucleic Acids Res. 36 (2008): 5678-5694.

[153]

A. Georgoulis, C. Vorgias, G. Chrousos, and E. Rogakou, “Genome Instability and gammaH2AX,” International Journal of Molecular Sciences 18 (2017): 1979.

[154]

C. Lucca, E. Ferrari, G. Shubassi, et al., “Sch9(S6K) controls DNA Repair and DNA Damage Response Efficiency in Aging Cells,” Cell reports 43 (2024): 114281.

[155]

S. Wang, X. Zhang, Y. Hou, et al., “SIRT6 activates PPARalpha to Improve Doxorubicin-induced Myocardial Cell Aging and Damage,” Chemico-Biological Interactions 392 (2024): 110920.

[156]

H. Kim, J. Cho, H. Quan, and J. Kim, “Down-regulation of Aurora B Kinase Induces Cellular Senescence in human Fibroblasts and Endothelial Cells Through a p53-dependent Pathway,” Febs Letters 585 (2011): 3569-3576.

[157]

B. Pierce, “The Aging Epigenome,” Elife 11 (2022): e78693.

[158]

B. Bartholomew, “Regulating the Chromatin Landscape: Structural and Mechanistic Perspectives,” Annual Review of Biochemistry 83 (2014): 671-696.

[159]

G. Cavalli and T. Misteli, “Functional Implications of Genome Topology,” Nature structural & molecular biology 20 (2013): 290-299.

[160]

B. Morris, B. Willcox, and T. Donlon, “Genetic and Epigenetic Regulation of human Aging and Longevity,” Biochim Biophys Acta Mol Basis Dis 1865 (2019): 1718-1744.

[161]

X. Wei, M. Murphy, N. Reddy, et al., “Redistribution of Lamina-associated Domains Reshapes Binding of Pioneer Factor FOXA2 in Development of Nonalcoholic Fatty Liver Disease,” Genome Research 32 (2022): 1981-1992.

[162]

E. Di Giorgio and L. Xodo, “Endogenous Retroviruses (ERVs): Does RLR (RIG-I-Like Receptors)-MAVS Pathway Directly Control Senescence and Aging as a Consequence of ERV De-Repression?” Frontiers in immunology 13 (2022): 917998.

[163]

M. Lachner, D. O'Carroll, S. Rea, K. Mechtler, and T. Jenuwein, “Methylation of Histone H3 Lysine 9 Creates a Binding Site for HP1 Proteins,” Nature 410 (2001): 116-120.

[164]

A. Bannister, P. Zegerman, J. Partridge, et al., “Selective Recognition of Methylated Lysine 9 on Histone H3 by the HP1 Chromo Domain,” Nature 410 (2001): 120-124.

[165]

C. Maison and G. Almouzni, “HP1 and the Dynamics of Heterochromatin Maintenance,” Nature Reviews Molecular Cell Biology 5 (2004): 296-304.

[166]

W. Zeng, A. Ball, and K. Yokomori, “HP1: Heterochromatin Binding Proteins Working the Genome,” Epigenetics 5 (2010): 287-292.

[167]

J. Padeken, S. Methot, and S. Gasser, “Establishment of H3K9-methylated Heterochromatin and Its Functions in Tissue Differentiation and Maintenance,” Nature Reviews Molecular Cell Biology 23 (2022): 623-640.

[168]

R. Li, Y. Teng, Y. Guo, et al., “Aging-related Decrease of Histone Methyltransferase SUV39H1 in Adipose-derived Stem Cells Enhanced SASP,” Mechanisms of Ageing and Development 215 (2023): 111868.

[169]

W. Zhang, J. Li, K. Suzuki, et al., “Aging Stem Cells. A Werner syndrome Stem Cell Model Unveils Heterochromatin Alterations as a Driver of human Aging,” Science 348 (2015): 1160-1163.

[170]

Z. Li, S. Duan, X. Hua, et al., “Asymmetric Distribution of Parental H3K9me3 in S Phase Silences L1 Elements,” Nature 623 (2023): 643-651.

[171]

G. Andrey and S. Mundlos, “The Three-dimensional Genome: Regulating Gene Expression During Pluripotency and Development,” Development (Cambridge, England) 144 (2017): 3646-3658.

[172]

F. Ma, Y. Cao, H. Du, et al., “Three-dimensional Chromatin Reorganization Regulates B Cell Development During Ageing,” Nature Cell Biology 26 (2024): 991-1002.

[173]

L. Bosch-Presegue and A. Vaquero, “Sirtuin-dependent Epigenetic Regulation in the Maintenance of Genome Integrity,” Febs Journal 282 (2015): 1745-1767.

[174]

Q. Gao, F. Chen, L. Zhang, et al., “Inhibition of DNA Methyltransferase Aberrations Reinstates Antioxidant Aging Suppressors and Ameliorates Renal Aging,” Aging Cell 21 (2022): e13526.

[175]

E. Pasyukova, A. Symonenko, O. Rybina, and A. Vaiserman, “Epigenetic Enzymes: A Role in Aging and Prospects for Pharmacological Targeting,” Ageing Research Reviews 67 (2021): 101312.

[176]

W. Li, W. Tian, G. Yuan, et al., “Molecular Basis of Nucleosomal H3K36 Methylation by NSD Methyltransferases,” Nature 590 (2021): 498-503.

[177]

Z. Li, X. Zhang, S. Xie, et al., “H3K36me2 methyltransferase NSD2 Orchestrates Epigenetic Reprogramming During Spermatogenesis,” Nucleic Acids Res. 50 (2022): 6786-6800.

[178]

S. Xiong, J. Zhou, and W. Chng, “Deciphering the Dynamics of Histone Acetylation and Chromatin Remodeling in Multiple Myeloma: A Tale Beyond the Tails,” Blood (2025): blood/202502840, https://doi.org/10.1182/blood.2025028403.

[179]

A. Pashos, A. Meyer, C. Bussey-Sutton, et al., “H3K36 methylation Regulates Cell Plasticity and Regeneration in the Intestinal Epithelium,” Nature Cell Biology 27 (2025): 202-217.

[180]

Y. Huang, X. Yang, Y. Wang, et al., “ARID1A recruits GATA2 to Regulate the Senescence of Trophoblast Cells Under High-glucose Condition,” Placenta 158 (2024): 156-164.

[181]

Z. Li, S. Zhu, X. Chen, et al., “ARID1A suppresses Malignant Transformation of human Pancreatic Cells via Mediating Senescence-associated miR-503/CDKN2A Regulatory Axis,” Biochemical and Biophysical Research Communications 493 (2017): 1018-1025.

[182]

B. Cabot and R. Cabot, “Chromatin Remodeling in Mammalian Embryos,” Reproduction (Cambridge, England) 155 (2018): R147-R158.

[183]

P. Mittal and C. Roberts, “The SWI/SNF Complex in Cancer—biology, Biomarkers and Therapy,” Nature reviews Clinical oncology 17 (2020): 435-448.

[184]

X. Li, G. Zhu, and B. Zhao, “Chromatin Remodeling in Tissue Stem Cell Fate Determination,” Cell Regen 13 (2024): 18.

[185]

D. Zhu, X. Wu, J. Zhou, et al., “NuRD Mediates Mitochondrial Stress-induced Longevity via Chromatin Remodeling in Response to Acetyl-CoA Level,” Science Advances 6 (2020): eabb2529.

[186]

J. Basta and M. Rauchman, “The Nucleosome Remodeling and Deacetylase Complex in Development and Disease,” Transl Res 165 (2015): 36-47.

[187]

N. Golden, M. Foley, K. Kim Guisbert, and E. Guisbert, “Divergent Regulatory Roles of NuRD Chromatin Remodeling Complex Subunits GATAD2 and CHD4 in Caenorhabditis elegans,” Genetics 221 (2022): iyac046.

[188]

P. Canale, J. Campolo, A. Borghini, and M. Andreassi, “Long Telomeric Repeat-Containing RNA (TERRA): Biological Functions and Challenges in Vascular Aging and Disease,” Biomedicines 11 (2023): 3211.

[189]

G. Libertini, G. Corbi, and F. Nicola, “Importance and Meaning of TERRA Sequences for Aging Mechanisms,” Biochemistry (Mosc) 85 (2020): 1505-1517.

[190]

E. Mahmoudi and M. Cairns, “CircRNA and Ageing,” Sub-Cellular Biochemistry 102 (2023): 249-270.

[191]

Q. Hao, L. Wang, M. Zhang, Z. Wang, M. Li, and X. Gao, “Taurine Stimulates Protein Synthesis and Proliferation of C2C12 Myoblast Cells Through the PI3K-ARID4B-mTOR Pathway,” British Journal of Nutrition 128 (2022): 1875-1886.

[192]

D. Xu, H. Sas-Nowosielska, G. Donahue, et al., “Histone Acetylation in an Alzheimer's Disease Cell Model Promotes Homeostatic Amyloid-reducing Pathways,” Acta Neuropathol Commun 12 (2024): 3.

[193]

L. Shu, Y. Zhang, Q. Sun, H. Pan, J. Guo, and B. Tang, “SNCA REP1 and Parkinson's Disease,” Neuroscience Letters 682 (2018): 79-84.

[194]

A. Cherian, P. K, and A. Vijayaraghavan, “Parkinson's Disease—genetic Cause,” Current Opinion in Neurology 36 (2023): 292-301.

[195]

H. Luo, G. Zhu, M. Eshelman, et al., “HOTTIP-dependent R-loop Formation Regulates CTCF Boundary Activity and TAD Integrity in Leukemia,” Molecular Cell 82 (2022): 833-851.

[196]

N. Ebrahim, K. Shakirova, and E. Dashinimaev, “PDX1 is the Cornerstone of Pancreatic Beta-cell Functions and Identity,” Frontiers in Molecular Biosciences 9 (2022): 1091757.

[197]

M. Chen, Z. Zhang, J. Ke, et al., “Chaetocin Attenuates Atherosclerosis Progression and Inhibits Vascular Smooth Muscle Cell Phenotype Switching,” J Cardiovasc Transl Res 15 (2022): 1270-1282.

[198]

C. Chronis, P. Fiziev, B. Papp, et al., “Cooperative Binding of Transcription Factors Orchestrates Reprogramming,” Cell 168 (2017): 442-459. e420.

[199]

S. Horvath, E. Lacunza, M. Mallat, et al., “Cognitive Rejuvenation in Old Rats by Hippocampal OSKM Gene Therapy,” Geroscience 47 (2025): 809-823.

[200]

A. Farsetti, B. Illi, and C. Gaetano, “How Epigenetics Impacts on human Diseases,” Eur J Intern Med 114 (2023): 15-22.

[201]

A. Barral, G. Pozo, L. Ducrot, et al., “SETDB1/NSD-dependent H3K9me3/H3K36me3 Dual Heterochromatin Maintains Gene Expression Profiles by Bookmarking Poised Enhancers,” Molecular Cell 82 (2022): 816-832. e812.

[202]

A. Korotkov, A. Seluanov, and V. Gorbunova, “Sirtuin 6: Linking Longevity With Genome and Epigenome Stability,” Trends in Cell Biology 31 (2021): 994-1006.

[203]

T. Hou, Z. Cao, J. Zhang, et al., “SIRT6 coordinates With CHD4 to Promote Chromatin Relaxation and DNA Repair,” Nucleic Acids Res. 48 (2020): 2982-3000.

[204]

R. Cai, R. Lv, X. Shi, G. Yang, and J. Jin, “CRISPR/dCas9 Tools: Epigenetic Mechanism and Application in Gene Transcriptional Regulation,” International Journal of Molecular Sciences 24 (2023): 14865.

[205]

M. Rahman and T. Tollefsbol, “Targeting Cancer Epigenetics With CRISPR-dCAS9: Principles and Prospects,” Methods (San Diego, Calif.) 187 (2021): 77-91.

[206]

T. Thum, “Non-coding RNAs in Ageing,” Ageing Research Reviews 17 (2014): 1-2.

[207]

M. Moqri, C. Herzog, J. Poganik, et al., “Biomarkers of Aging for the Identification and Evaluation of Longevity Interventions,” Cell 186 (2023): 3758-3775.

[208]

W. Lai, M. Lin, and W. Wong, “Tackling Aging by Using miRNA as a Target and a Tool,” Trends in Molecular Medicine 25 (2019): 673-684.

[209]

J. Wagner, L. Tombor, P. Malacarne, et al., “Aging Impairs the Neurovascular Interface in the Heart,” Science 381 (2023): 897-906.

[210]

V. Swahari, A. Nakamura, E. Hollville, et al., “miR-29 Is an Important Driver of Aging-related Phenotypes,” Communications Biology 7 (2024): 1055.

[211]

Y. Bi, X. Qiao, Z. Cai, et al., “Exosomal miR-302b Rejuvenates Aging Mice by Reversing the Proliferative Arrest of Senescent Cells,” Cell metabolism 37 (2025): 527-541. e526.

[212]

T. Smith-Vikos and F. Slack, “MicroRNAs and Their Roles in Aging,” Journal of Cell Science 125 (2012): 7-17.

[213]

X. Bai, Y. Ma, R. Ding, B. Fu, S. Shi, and X. Chen, “miR-335 and miR-34a Promote Renal Senescence by Suppressing Mitochondrial Antioxidative Enzymes,” Journal of the American Society of Nephrology 22 (2011): 1252-1261.

[214]

D. Bhaumik, G. Scott, S. Schokrpur, et al., “MicroRNAs miR-146a/b Negatively Modulate the Senescence-associated Inflammatory Mediators IL-6 and IL-8,” Aging (Albany NY) 1 (2009): 402-411.

[215]

S. Ghafouri-Fard, A. Abak, S. Talebi, et al., “Role of miRNA and lncRNAs in Organ Fibrosis and Aging,” Biomedicine & Pharmacotherapy 143 (2021): 112132.

[216]

Z. Yang, S. Jiang, J. Shang, et al., “LncRNA: Shedding Light on Mechanisms and Opportunities in Fibrosis and Aging,” Ageing Research Reviews 52 (2019): 17-31.

[217]

H. Gao, E. Nepovimova, Z. Heger, et al., “Role of Hypoxia in Cellular Senescence,” Pharmacological Research 194 (2023): 106841.

[218]

B. Wang, C. Suen, H. Ma, et al., “The Roles of H19 in Regulating Inflammation and Aging,” Frontiers in immunology 11 (2020): 579687.

[219]

L. Nasiri, M. Vaez-Mahdavi, H. Hassanpour, et al., “Transcription of Biological Aging Markers (ANRIL, P16(INK4a), TBX2, and TERRA) and Their Correlations With Severity of Sulfur Mustard Exposure in Veterans,” Drug and Chemical Toxicology (2024): 1-9, https://doi.org/10.1080/01480545.2024.2395571.

[220]

Y. Kotake, T. Nakagawa, K. Kitagawa, et al., “Long Non-coding RNA ANRIL Is Required for the PRC2 Recruitment to and Silencing of p15(INK4B) Tumor Suppressor Gene,” Oncogene 30 (2011): 1956-1962.

[221]

S. Hu, J. Zheng, Z. Du, and G. Wu, “Knock Down of lncRNA H19 Promotes Axon Sprouting and Functional Recovery After Cerebral Ischemic Stroke,” Brain Research 1732 (2020): 146681.

[222]

C. Ma, K. Nong, H. Zhu, et al., “H19 promotes Pancreatic Cancer Metastasis by Derepressing let-7's Suppression on Its Target HMGA2-mediated EMT,” Tumour Biology: The Journal of the International Society for Oncodevelopmental Biology and Medicine 35 (2014): 9163-9169.

[223]

H. Gruner, M. Cortes-Lopez, D. Cooper, M. Bauer, and P. Miura, “CircRNA Accumulation in the Aging Mouse Brain,” Scientific Reports 6 (2016): 38907.

[224]

D. Knupp and P. Miura, “CircRNA Accumulation: A New Hallmark of Aging?” Mechanisms of Ageing and Development 173 (2018): 71-79.

[225]

L. Zhang, Y. Zhang, F. Yu, X. Li, H. Gao, and P. Li, “The circRNA-miRNA/RBP Regulatory Network in Myocardial Infarction,” Frontiers in pharmacology 13 (2022): 941123.

[226]

N. Liu, Z. Zhang, Y. Wu, Y. Wang, and Y. Liang, “CRBSP:Prediction of CircRNA-RBP Binding Sites Based on Multimodal Intermediate Fusion,” IEEE/ACM Trans Comput Biol Bioinform 20 (2023): 2898-2906.

[227]

F. Ding, L. Lu, C. Wu, et al., “circHIPK3 prevents Cardiac Senescence by Acting as a Scaffold to Recruit Ubiquitin Ligase to Degrade HuR,” Theranostics 12 (2022): 7550-7566.

[228]

Y. Xu, Z. Zai, Z. Lu, et al., “Circular RNA CircCDKN2B-AS_006 Promotes the Tumor-Like Growth and Metastasis of Rheumatoid Arthritis Synovial Fibroblasts by Targeting the miR-1258/RUNX1 Axis,” International Journal of Molecular Sciences 24 (2023): 5880.

[229]

C. Zhao, X. Li, G. Sun, et al., “CircFOXO3 protects Against Osteoarthritis by Targeting Its Parental Gene FOXO3 and Activating PI3K/AKT-mediated Autophagy,” Cell death & disease 13 (2022): 932.

[230]

Y. Zhang, W. Zhu, F. Qi, and F. Che, “CircHIPK3 promotes Neuroinflammation Through Regulation of the miR-124-3p/STAT3/NLRP3 Signaling Pathway in Parkinson's Disease,” Advances in clinical and experimental medicine: official organ Wroclaw Medical University 32 (2023): 315-329.

[231]

E. Lecona, L. Rojas, R. Bonasio, A. Johnston, O. Fernandez-Capetillo, and D. Reinberg, “Polycomb Protein SCML2 Regulates the Cell Cycle by Binding and Modulating CDK/CYCLIN/p21 Complexes,” Plos Biology 11 (2013): e1001737.

[232]

W. Du, W. Yang, E. Liu, Z. Yang, P. Dhaliwal, and B. Yang, “Foxo3 circular RNA Retards Cell Cycle Progression via Forming Ternary Complexes With p21 and CDK2,” Nucleic Acids Res. 44 (2016): 2846-2858.

[233]

A. Seyhan, “Trials and Tribulations of MicroRNA Therapeutics,” International Journal of Molecular Sciences 25 (2024): 1469.

[234]

S. Anver, A. Sumit, X. Sun, et al., “Ageing-associated Long Non-coding RNA Extends Lifespan and Reduces Translation in Non-dividing Cells,” Embo Reports 25 (2024): 4921-4949.

[235]

W. Schain, “Psychosocial Issues in Breast Cancer Clinical Trials,” Recent Results in Cancer Research 127 (1993): 235-241.

[236]

M. Fitz-James and G. Cavalli, “Molecular Mechanisms of Transgenerational Epigenetic Inheritance,” Nature Reviews Genetics 23 (2022): 325-341.

[237]

M. Xavier, S. Roman, R. Aitken, and B. Nixon, “Transgenerational Inheritance: How Impacts to the Epigenetic and Genetic Information of Parents Affect Offspring Health,” Human Reproduction Update 25 (2019): 518-540.

[238]

K. Moelling, “Epigenetics and Transgenerational Inheritance,” The Journal of Physiology 602 (2024): 2537-2545.

[239]

E. Lister-Shimauchi, B. McCarthy, M. Lippincott, and S. Ahmed, “Genetic and Epigenetic Inheritance at Telomeres,” Epigenomes 6 (2022): 9.

[240]

M. Haberman, T. Menashe, N. Cohen, et al., “Paternal High-fat Diet Affects Weight and DNA Methylation of Their Offspring,” Scientific Reports 14 (2024): 19874.

[241]

Q. Zhang, X. Xiao, J. Zheng, et al., “Maternal High-Fat Diet Disturbs the DNA Methylation Profile in the Brown Adipose Tissue of Offspring Mice,” Front Endocrinol (Lausanne) 12 (2021): 705827.

[242]

L. Oblak, J. van der Zaag, A. Higgins-Chen, M. Levine, and M. Boks, “A Systematic Review of Biological, Social and Environmental Factors Associated With Epigenetic Clock Acceleration,” Ageing Research Reviews 69 (2021): 101348.

[243]

P. Anjaria, V. Asediya, J. Nayak, and P. Koringa, “The Epigenetic Landscape: How Environmental Cues Shape Gene Expression,” Epigenomics 15 (2023): 267-270.

[244]

G. Lidzbarsky, D. Gutman, H. Shekhidem, L. Sharvit, and G. Atzmon, “Genomic Instabilities, Cellular Senescence, and Aging: In Vitro, in Vivo and Aging-Like Human Syndromes,” Front Med (Lausanne) 5 (2018): 104.

[245]

A. Brooks-Wilson, “Genetics of Healthy Aging and Longevity,” Human Genetics 132 (2013): 1323-1338.

[246]

M. Petr, T. Tulika, L. Carmona-Marin, and M. Scheibye-Knudsen, “Protecting the Aging Genome,” Trends in Cell Biology 30 (2020): 117-132.

[247]

A. Aguilera and B. Gomez-Gonzalez, “Genome Instability: A Mechanistic View of Its Causes and Consequences,” Nature Reviews Genetics 9 (2008): 204-217.

[248]

A. Aguilera and T. Garcia-Muse, “Causes of Genome Instability,” Annual Review of Genetics 47 (2013): 1-32.

[249]

V. Gorbunova, A. Seluanov, Z. Mao, and C. Hine, “Changes in DNA Repair During Aging,” Nucleic Acids Res. 35 (2007): 7466-7474.

[250]

A. Tubbs and A. Nussenzweig, “Endogenous DNA Damage as a Source of Genomic Instability in Cancer,” Cell 168 (2017): 644-656.

[251]

J. Ruszkiewicz, A. Burkle, and A. Mangerich, “Fueling Genome Maintenance: On the Versatile Roles of NAD(+) in Preserving DNA Integrity,” Journal of Biological Chemistry 298 (2022): 102037.

[252]

R. Prakash, Y. Zhang, W. Feng, and M. Jasin, “Homologous Recombination and human Health: The Roles of BRCA1, BRCA2, and Associated Proteins,” Cold Spring Harbor perspectives in biology 7 (2015): a016600.

[253]

K. Karakostis, L. Malbert-Colas, A. Thermou, B. Vojtesek, and R. Fahraeus, “The DNA Damage Sensor ATM Kinase Interacts With the p53 mRNA and Guides the DNA Damage Response Pathway,” Molecular cancer 23 (2024): 21.

[254]

Z. Mirman, K. Sharma, T. Carroll, and T. de Lange, “Expression of BRCA1, BRCA2, RAD51, and Other DSB Repair Factors Is Regulated by CRL4(WDR70),” Dna Repair 113 (2022): 103320.

[255]

E. Fang, H. Kassahun, D. Croteau, et al., “NAD(+) Replenishment Improves Lifespan and Healthspan in Ataxia Telangiectasia Models via Mitophagy and DNA Repair,” Cell metabolism 24 (2016): 566-581.

[256]

A. Wilk, F. Hayat, R. Cunningham, et al., “Extracellular NAD(+) Enhances PARP-dependent DNA Repair Capacity Independently of CD73 Activity,” Scientific Reports 10 (2020): 651.

[257]

J. Alhmoud, J. Woolley, A. Al Moustafa, and M. Malki, “DNA Damage/Repair Management in Cancers,” Cancers (Basel) 12 (2020): 1050.

[258]

J. He, X. Fu, M. Zhang, et al., “Transposable Elements Are Regulated by Context-specific Patterns of Chromatin Marks in Mouse Embryonic Stem Cells,” Nature Communications 10 (2019): 34.

[259]

A. Fernandez, C. O'Leary, K. O'Byrne, J. Burgess, D. Richard, and A. Suraweera, “Epigenetic Mechanisms in DNA Double Strand Break Repair: A Clinical Review,” Frontiers in Molecular Biosciences 8 (2021): 685440.

[260]

R. Kumari and P. Jat, “Mechanisms of Cellular Senescence: Cell Cycle Arrest and Senescence Associated Secretory Phenotype,” Frontiers in Cell and Developmental Biology 9 (2021): 645593.

[261]

Y. Qin, H. Liu, and H. Wu, “Cellular Senescence in Health, Disease, and Lens Aging,” Pharmaceuticals (Basel) 18 (2025): 244.

[262]

R. Tenchov, J. Sasso, X. Wang, and Q. Zhou, “Aging Hallmarks and Progression and Age-Related Diseases: A Landscape View of Research Advancement,” Acs Chemical Neuroscience 15 (2024): 1-30.

[263]

S. He and N. Sharpless, “Senescence in Health and Disease,” Cell 169 (2017): 1000-1011.

[264]

X. Huang, L. Huang, J. Lu, et al., “The Relationship Between Telomere Length and Aging-related Diseases,” Clinical and Experimental Medicine 25 (2025): 72.

[265]

J. de Magalhaes and J. Passos, “Stress, Cell Senescence and Organismal Ageing,” Mechanisms of Ageing and Development 170 (2018): 2-9.

[266]

R. Benetti, M. Garcia-Cao, and M. Blasco, “Telomere Length Regulates the Epigenetic Status of Mammalian Telomeres and Subtelomeres,” Nature Genetics 39 (2007): 243-250.

[267]

S. Gadalla, H. Katki, F. Shebl, N. Giri, B. Alter, and S. Savage, “The Relationship Between DNA Methylation and Telomere Length in Dyskeratosis Congenita,” Aging Cell 11 (2012): 24-28.

[268]

L. Chen, C. Zhang, W. Ma, J. Huang, Y. Zhao, and H. Liu, “METTL3-mediated m6A Modification Stabilizes TERRA and Maintains Telomere Stability,” Nucleic Acids Res. 50 (2022): 11619-11634.

[269]

H. Tang, H. Wang, X. Cheng, et al., “HuR Regulates Telomerase Activity Through TERC Methylation,” Nature Communications 9 (2018): 2213.

[270]

X. Cheng, X. Gu, T. Xia, et al., “HuB and HuD Repress Telomerase Activity by Dissociating HuR From TERC,” Nucleic Acids Res. 49 (2021): 2848-2858.

[271]

J. Rivosecchi, K. Jurikova, and E. Cusanelli, “Telomere-specific Regulation of TERRA and Its Impact on Telomere Stability,” Seminars in cell & developmental biology 157 (2024): 3-23.

[272]

F. Milosic, M. Hengstschlager, and S. Osmanagic-Myers, “Premature Aging in Genetic Diseases: What Conclusions Can be Drawn for Physiological Aging,” Front Aging 4 (2023): 1327833.

[273]

S. Maynard, E. Fang, M. Scheibye-Knudsen, D. Croteau, and V. Bohr, “DNA Damage, DNA Repair, Aging, and Neurodegeneration,” Cold Spring Harbor perspectives in medicine 5 (2015): a025130.

[274]

M. Li, H. Guo, M. Carey, and C. Huang, “Transcriptional and Epigenetic Dysregulation Impairs Generation of Proliferative Neural Stem and Progenitor Cells During Brain Aging,” Nat Aging 4 (2024): 62-79.

[275]

G. Lupo, P. Nisi, P. Esteve, et al., “Molecular Profiling of Aged Neural Progenitors Identifies Dbx2 as a Candidate Regulator of Age-associated Neurogenic Decline,” Aging Cell 17 (2018): e12745.

[276]

J. Wood, B. Jones, N. Jiang, et al., “Chromatin-modifying Genetic Interventions Suppress Age-associated Transposable Element Activation and Extend Life Span in Drosophila,” PNAS 113 (2016): 11277-11282.

[277]

A. Du, J. Chobirko, X. Zhuo, C. Feschotte, and T. Wang, “Regulatory Transposable Elements in the Encyclopedia of DNA Elements,” Nature Communications 15 (2024): 7594.

[278]

C. Mrabti, N. Yang, G. Desdin-Mico, et al., “Loss of H3K9 Trimethylation Leads to Premature Aging,” BioRxiv (2024), https://doi.org/10.1101/2024.07.24.604929.

[279]

B. Benayoun, E. Pollina, and A. Brunet, “Epigenetic Regulation of Ageing: Linking Environmental Inputs to Genomic Stability,” Nature Reviews Molecular Cell Biology 16 (2015): 593-610.

[280]

A. Gebrie, “Transposable Elements as Essential Elements in the Control of Gene Expression,” Mob DNA 14 (2023): 9.

[281]

C. Ravel-Godreuil, R. Znaidi, T. Bonnifet, R. Joshi, and J. Fuchs, “Transposable Elements as New Players in Neurodegenerative Diseases,” Febs Letters 595 (2021): 2733-2755.

[282]

F. Della Valle, P. Reddy, A. Aguirre Vazquez, and J. Izpisua Belmonte, “Reactivation of Retrotransposable Elements Is Associated With Environmental Stress and Ageing,” Nature Reviews Genetics 26 (2025): 547-558.

[283]

B. McCauley, L. Sun, R. Yu, et al., “Altered Chromatin States Drive Cryptic Transcription in Aging Mammalian Stem Cells,” Nat Aging 1 (2021): 684-697.

[284]

C. Li, K. Chen, X. Li, and X. Xiong, “Epitranscriptome-epigenome Interactions in Development and Disease Mechanisms,” Trends in Genetics 41, no. 8 (2025): 691-705, https://doi.org/10.1016/j.tig.2025.04.009.

[285]

M. Schosserer, N. Minois, T. Angerer, et al., “Corrigendum: Methylation of Ribosomal RNA by NSUN5 Is a Conserved Mechanism Modulating Organismal Lifespan,” Nature Communications 7 (2016): 11530.

[286]

K. Lei, S. Lin, and Q. Yuan, “N6-methyladenosine (m6A) Modification of Ribosomal RNAs (rRNAs): Critical Roles in mRNA Translation and Diseases,” Genes Dis 10 (2023): 126-134.

[287]

X. He, S. Memczak, J. Qu, J. Belmonte, and G. Liu, “Single-cell Omics in Ageing: A Young and Growing Field,” Nat Metab 2 (2020): 293-302.

[288]

A. Papantonis, A. Antebi, L. Partridge, and A. Beyer, “Age-associated Changes in Transcriptional Elongation and Their Effects on Homeostasis,” Trends in Cell Biology 35, no. 8 (2024): 645-650, https://doi.org/10.1016/j.tcb.2024.11.005.

[289]

J. Neves, P. Sousa-Victor, and H. Jasper, “Rejuvenating Strategies for Stem Cell-Based Therapies in Aging,” Cell Stem Cell 20 (2017): 161-175.

[290]

D. Saavedra, A. Ane-Kouri, N. Barzilai, et al., “Aging and Chronic Inflammation: Highlights From a Multidisciplinary Workshop,” Immun Ageing 20 (2023): 25.

[291]

B. Liu, J. Qu, W. Zhang, J. Izpisua Belmonte, and G. Liu, “A Stem Cell Aging Framework, From Mechanisms to Interventions,” Cell reports 41 (2022): 111451.

[292]

A. Ahmed, M. Sheng, S. Wasnik, D. Baylink, and K. Lau, “Effect of Aging on Stem Cells,” World J Exp Med 7 (2017): 1-10.

[293]

C. Chen, L. Zhang, R. Dutta, et al., “SRCAP Mutations Drive Clonal Hematopoiesis Through Epigenetic and DNA Repair Dysregulation,” Cell Stem Cell 30 (2023): 1503-1519. e1508.

[294]

R. Yan, X. Cheng, C. Gu, et al., “Dynamics of DNA Hydroxymethylation and Methylation During Mouse Embryonic and Germline Development,” Nature Genetics 55 (2023): 130-143.

[295]

H. Blau, B. Cosgrove, and A. Ho, “The central Role of Muscle Stem Cells in Regenerative Failure With Aging,” Nature Medicine 21 (2015): 854-862.

[296]

Z. Chen, M. Rasheed, and Y. Deng, “The Epigenetic Mechanisms Involved in Mitochondrial Dysfunction: Implication for Parkinson's Disease,” Brain Pathology 32 (2022): e13012.

[297]

R. Wang, H. Sun, G. Wang, and H. Ren, “Imbalance of Lysine Acetylation Contributes to the Pathogenesis of Parkinson's Disease,” International Journal of Molecular Sciences 21 (2020): 7182.

[298]

X. Gao, J. Ding, J. Xie, and H. Xu, “Epigenetic Regulation of Iron Metabolism and Ferroptosis in Parkinson's disease: Identifying Novel Epigenetic Targets,” Acta Pharmacologica Sinica 46 (2025): 2075-2092.

[299]

M. Vermulst, J. Bielas, G. Kujoth, et al., “Mitochondrial Point Mutations Do Not Limit the Natural Lifespan of Mice,” Nature Genetics 39 (2007): 540-543.

[300]

A. Trifunovic, A. Wredenberg, M. Falkenberg, et al., “Premature Ageing in Mice Expressing Defective Mitochondrial DNA Polymerase,” Nature 429 (2004): 417-423.

[301]

E. Lesnefsky, Q. Chen, and C. Hoppel, “Mitochondrial Metabolism in Aging Heart,” Circulation Research 118 (2016): 1593-1611.

[302]

M. Falk, “Neurodevelopmental Manifestations of Mitochondrial Disease,” Journal of Developmental and Behavioral Pediatrics 31 (2010): 610-621.

[303]

G. Zhang, H. Wei, A. Zhao, et al., “Mitochondrial DNA Leakage: Underlying Mechanisms and Therapeutic Implications in Neurological Disorders,” J Neuroinflammation 22 (2025): 34.

[304]

M. Yan, Y. Li, Q. Luo, et al., “Mitochondrial Damage and Activation of the Cytosolic DNA Sensor cGAS-STING Pathway Lead to Cardiac Pyroptosis and Hypertrophy in Diabetic Cardiomyopathy Mice,” Cell Death Discov 8 (2022): 258.

[305]

C. Wiley, M. Velarde, P. Lecot, et al., “Mitochondrial Dysfunction Induces Senescence With a Distinct Secretory Phenotype,” Cell metabolism 23 (2016): 303-314.

[306]

H. Zhang, D. Ryu, Y. Wu, et al., “NAD(+) Repletion Improves Mitochondrial and Stem Cell Function and Enhances Life Span in Mice,” Science 352 (2016): 1436-1443.

[307]

S. Stanciu, M. Jinga, D. Miricescu, et al., “mTOR Dysregulation, Insulin Resistance, and Hypertension,” Biomedicines 12 (2024): 1802.

[308]

D. Wang, D. Ji, C. Yu, D. Wu, and L. Qi, “Research Progress on the Mitochondrial Mechanism of Age-related Non-alcoholic Fatty Liver,” World Journal of Gastroenterology 29 (2023): 1982-1993.

[309]

J. Kim, Y. Wei, and J. Sowers, “Role of Mitochondrial Dysfunction in Insulin Resistance,” Circulation Research 102 (2008): 401-414.

[310]

R. Naik, M. Mir, I. Malik, et al., “The Potential Mechanism and the Role of Antioxidants in Mitigating Oxidative Stress in Alzheimer's Disease,” Front Biosci (Landmark Ed) 30 (2025): 25551.

[311]

A. Terman, H. Dalen, J. Eaton, J. Neuzil, and U. Brunk, “Aging of Cardiac Myocytes in Culture: Oxidative Stress, Lipofuscin Accumulation, and Mitochondrial Turnover,” Ann N Y Acad Sci 1019 (2004): 70-77.

[312]

J. Peoples, A. Saraf, N. Ghazal, T. Pham, and J. Kwong, “Mitochondrial Dysfunction and Oxidative Stress in Heart Disease,” Experimental & Molecular Medicine 51 (2019): 1-13.

[313]

K. Cao, K. Wang, M. Yang, X. Liu, W. Lv, and J. Liu, “Punicalagin Improves Hepatic Lipid Metabolism via Modulation of Oxidative Stress and Mitochondrial Biogenesis in Hyperlipidemic Mice,” Food Funct 11 (2020): 9624-9633.

[314]

R. Kanwal and S. Gupta, “Epigenetic Modifications in Cancer,” Clinical Genetics 81 (2012): 303-311.

[315]

A. Eden, F. Gaudet, A. Waghmare, and R. Jaenisch, “Chromosomal Instability and Tumors Promoted by DNA Hypomethylation,” Science 300 (2003): 455.

[316]

C. Nguyen, G. Liang, T. Nguyen, et al., “Susceptibility of Nonpromoter CpG Islands to De Novo Methylation in Normal and Neoplastic Cells,” JNCI: Journal of the National Cancer Institute 93 (2001): 1465-1472.

[317]

L. Borkiewicz, “Histone 3 Lysine 27 Trimethylation Signature in Breast Cancer,” International Journal of Molecular Sciences 22 (2021): 12853.

[318]

G. Taylor, R. Eskeland, B. Hekimoglu-Balkan, M. Pradeepa, and W. Bickmore, “H4K16 acetylation Marks Active Genes and Enhancers of Embryonic Stem Cells, but Does Not Alter Chromatin Compaction,” Genome Research 23 (2013): 2053-2065.

[319]

P. Das and J. Taube, “Regulating Methylation at H3K27: A Trick or Treat for Cancer Cell Plasticity,” Cancers (Basel) 12 (2020): 2792.

[320]

Y. Peng, X. Zhang, X. Feng, X. Fan, and Z. Jin, “The Crosstalk Between microRNAs and the Wnt/Beta-catenin Signaling Pathway in Cancer,” Oncotarget 8 (2017): 14089-14106.

[321]

H. Qian, M. Maghsoudloo, P. Kaboli, et al., “Decoding the Promise and Challenges of miRNA-Based Cancer Therapies: An Essential Update on miR-21, miR-34, and miR-155,” International journal of medical sciences 21 (2024): 2781-2798.

[322]

W. Zhang, K. Zhang, J. Shi, et al., “The Impact of the Senescent Microenvironment on Tumorigenesis: Insights for Cancer Therapy,” Aging Cell 23 (2024): e14182.

[323]

A. Suraweera, K. O'Byrne, and D. Richard, “Epigenetic Drugs in Cancer Therapy,” Cancer and Metastasis Reviews 44 (2025): 37.

[324]

H. Hampel, R. Vassar, B. De Strooper, et al., “The Beta-Secretase BACE1 in Alzheimer's Disease,” Biological Psychiatry 89 (2021): 745-756.

[325]

L. Lossi, C. Castagna, and A. Merighi, “An Overview of the Epigenetic Modifications in the Brain Under Normal and Pathological Conditions,” International Journal of Molecular Sciences 25 (2024): 3881.

[326]

M. Caillet-Boudin, L. Buee, N. Sergeant, and B. Lefebvre, “Regulation of human MAPT Gene Expression,” Mol Neurodegener 10 (2015): 28.

[327]

H. Mori, Y. Yoshino, M. Ueno, et al., “Blood MAPT Expression and Methylation Status in Alzheimer's Disease,” PCN Rep 1 (2022): e65.

[328]

L. de Mena, G. Pares, A. Garrido, et al., “alpha-Synuclein Gene Hypomethylation in LRRK2 Parkinson's Disease Patients,” Movement Disorders 40 (2025): 550-555.

[329]

L. Wang, L. Liu, C. Han, et al., “Histone Deacetylase 4 Inhibition Reduces Rotenone-Induced Alpha-Synuclein Accumulation via Autophagy in SH-SY5Y Cells,” Brain Sci 13 (2023): 670.

[330]

R. Wierda, S. Geutskens, J. Jukema, P. Quax, and P. van den Elsen, “Epigenetics in Atherosclerosis and Inflammation,” Journal of Cellular and Molecular Medicine 14 (2010): 1225-1240.

[331]

X. Chen, Y. He, W. Fu, et al., “Histone Deacetylases (HDACs) and Atherosclerosis: A Mechanistic and Pharmacological Review,” Frontiers in Cell and Developmental Biology 8 (2020): 581015.

[332]

J. Nikolajevic, N. Ariaee, A. Liew, S. Abbasnia, B. Fazeli, and M. Sabovic, “The Role of MicroRNAs in Endothelial Cell Senescence,” Cells 11 (2022): 1185.

[333]

M. Yamakuchi and T. Hashiguchi, “Endothelial Cell Aging: How miRNAs Contribute?” Journal of Clinical Medicine 7 (2018): 170.

[334]

K. Crider, T. Yang, R. Berry, and L. Bailey, “Folate and DNA Methylation: A Review of Molecular Mechanisms and the Evidence for Folate's Role,” Advances in nutrition 3 (2012): 21-38.

[335]

S. Ahmed, S. Ansari, E. Mensah-Brown, and B. Emerald, “The Role of DNA Methylation in the Pathogenesis of Type 2 Diabetes Mellitus,” Clin Epigenetics 12 (2020): 104.

[336]

S. Klimczak and A. Sliwinska, “Epigenetic Regulation of Inflammation in Insulin Resistance,” Seminars in cell & developmental biology 154 (2024): 185-192.

[337]

H. Ibrahim, “Epigenetic Regulation of Obesity-Associated Type 2 Diabetes,” Medicina (Kaunas, Lithuania) 58 (2022): 1028-1044.

[338]

C. Jazieh, T. Arabi, Z. Asim, et al., “Unraveling the Epigenetic Fabric of Type 2 Diabetes Mellitus: Pathogenic Mechanisms and Therapeutic Implications,” Front Endocrinol (Lausanne) 15 (2024): 1295967.

[339]

M. Zaiou, “Peroxisome Proliferator-Activated Receptor-gamma as a Target and Regulator of Epigenetic Mechanisms in Nonalcoholic Fatty Liver Disease,” Cells 12 (2023): 1205.

[340]

S. Wang, L. Zha, X. Cui, et al., “Epigenetic Regulation of Hepatic Lipid Metabolism by DNA Methylation,” Adv Sci (Weinh) 10 (2023): e2206068.

[341]

Y. Shi and W. Qi, “Histone Modifications in NAFLD: Mechanisms and Potential Therapy,” International Journal of Molecular Sciences 24 (2023): 14653.

[342]

S. Pillai, H. Lakhani, M. Zehra, et al., “Predicting Nonalcoholic Fatty Liver Disease Through a Panel of Plasma Biomarkers and MicroRNAs in Female West Virginia Population,” International Journal of Molecular Sciences 21 (2020): 6698.

[343]

J. Shen, X. Lin, F. Dai, et al., “Ubiquitin-specific Peptidases: Players in Bone Metabolism,” Cell Proliferation 56 (2023): e13444.

[344]

L. Hu, W. Chen, A. Qian, and Y. Li, “Wnt/Beta-catenin Signaling Components and Mechanisms in Bone Formation, Homeostasis, and Disease,” Bone Res 12 (2024): 39.

[345]

Y. Zhang, X. Feng, B. Zheng, and Y. Liu, “Regulation and Mechanistic Insights Into Tensile Strain in Mesenchymal Stem Cell Osteogenic Differentiation,” Bone 187 (2024): 117197.

[346]

L. Zhang, Q. Guan, Z. Wang, J. Feng, J. Zou, and B. Gao, “Consequences of Aging on Bone,” Aging Dis 15 (2023): 2417-2452.

[347]

J. Taylor, J. Wood, E. Mizerak, et al., “Sirt6 regulates Lifespan in Drosophila Melanogaster,” PNAS 119 (2022): e2111176119.

[348]

W. Yu, H. Wang, J. Zhang, and C. Yin, “The Effects of Epigenetic Modifications on Bone Remodeling in Age-related Osteoporosis,” Connective Tissue Research 64 (2023): 105-116.

[349]

K. Park-Min, “Epigenetic Regulation of Bone Cells,” Connective Tissue Research 58 (2017): 76-89.

[350]

K. Takahashi and S. Yamanaka, “Induction of Pluripotent Stem Cells From Mouse Embryonic and Adult Fibroblast Cultures by Defined Factors,” Cell 126 (2006): 663-676.

[351]

P. Mehrotra, J. Jablonski, J. Toftegaard, et al., “Skeletal Muscle Reprogramming Enhances Reinnervation After Peripheral Nerve Injury,” Nature Communications 15 (2024): 9218.

[352]

V. Tabar, H. Sarva, A. Lozano, et al., “Phase I Trial of hES Cell-derived Dopaminergic Neurons for Parkinson's disease,” Nature 641, no. 8064 (2025): 978-983.

[353]

N. Sawamoto, D. Doi, E. Nakanishi, et al., “Phase I/II Trial of iPS-cell-derived Dopaminergic Cells for Parkinson's disease,” Nature 641, no. 8064 (2025): 971-977, https://doi.org/10.1038/s41586-025-08700-0.

[354]

L. Pozzo, Z. Xu, S. Lin, et al., “Role of Epigenetics in the Regulation of Skin Aging and Geroprotective Intervention: A New Sight,” Biomedicine & Pharmacotherapy 174 (2024): 116592.

[355]

M. Jeong, Y. Lin, S. Wennersten, et al., “Histone Deacetylase Activity Governs Diastolic Dysfunction Through a Nongenomic Mechanism,” Science Translational Medicine 10 (2018): eaao0144.

[356]

M. Walsh, A. Bhattacharya, K. Sataranatarajan, et al., “The Histone Deacetylase Inhibitor Butyrate Improves Metabolism and Reduces Muscle Atrophy During Aging,” Aging Cell 14 (2015): 957-970.

[357]

S. Huang, H. Shi, Z. Shi, J. Wu, and L. He, “Vorinostat, a Potential Hormetin, Extends Lifespan and Enhances Stress Resistance via the SKN-1 Pathway in Caenorhabditis elegans,” Biogerontology 26 (2025): 97.

[358]

C. Falckenhayn, A. Bienkowska, J. Sohle, et al., “Identification of Dihydromyricetin as a Natural DNA Methylation Inhibitor With Rejuvenating Activity in human Skin,” Front Aging 4 (2023): 1258184.

[359]

S. Yamaguchi, J. Irie, M. Mitsuishi, et al., “Safety and Efficacy of Long-term Nicotinamide Mononucleotide Supplementation on Metabolism, Sleep, and Nicotinamide Adenine Dinucleotide Biosynthesis in Healthy, Middle-aged Japanese Men,” Endocrine Journal 71 (2024): 153-169.

[360]

K. Pencina, R. Valderrabano, B. Wipper, et al., “Nicotinamide Adenine Dinucleotide Augmentation in Overweight or Obese Middle-Aged and Older Adults: A Physiologic Study,” Journal of Clinical Endocrinology and Metabolism 108 (2023): 1968-1980.

[361]

Y. Yang, X. Lu, N. Liu, et al., “Metformin Decelerates Aging Clock in Male Monkeys,” Cell 187 (2024): 6358-6378. e6329.

[362]

J. Mannick and D. Lamming, “Targeting the Biology of Aging With mTOR Inhibitors,” Nat Aging 3 (2023): 642-660.

[363]

S. Horvath, A. Lu, H. Cohen, and K. Raj, “Rapamycin Retards Epigenetic Ageing of Keratinocytes Independently of Its Effects on Replicative Senescence, Proliferation and Differentiation,” Aging (Albany NY) 11 (2019): 3238-3249.

[364]

J. Zhai, W. Kongsberg, Y. Pan, C. Hao, X. Wang, and J. Sun, “Caloric Restriction Induced Epigenetic Effects on Aging,” Frontiers in Cell and Developmental Biology 10 (2022): 1079920.

[365]

D. Hernandez-Saavedra, L. Moody, G. Xu, H. Chen, and Y. Pan, “Epigenetic Regulation of Metabolism and Inflammation by Calorie Restriction,” Advances in nutrition 10 (2019): 520-536.

[366]

M. Wei, S. Brandhorst, M. Shelehchi, et al., “Fasting-mimicking Diet and Markers/Risk Factors for Aging, Diabetes, Cancer, and Cardiovascular Disease,” Science Translational Medicine 9 (2017): eaai8700.

[367]

W. Hastings, Q. Ye, S. Wolf, et al., “Effect of Long-term Caloric Restriction on Telomere Length in Healthy Adults: CALERIE 2 Trial Analysis,” Aging Cell 23 (2024): e14149.

[368]

Y. Kim, T. Huan, R. Joehanes, et al., “Higher Diet Quality Relates to Decelerated Epigenetic Aging,” American Journal of Clinical Nutrition 115 (2022): 163-170.

[369]

A. Yaskolka Meir, M. Keller, A. Hoffmann, et al., “The Effect of Polyphenols on DNA Methylation-assessed Biological Age Attenuation: The DIRECT PLUS Randomized Controlled Trial,” BMC Medicine [Electronic Resource] 21 (2023): 364.

[370]

V. Dwaraka, L. Aronica, N. Carreras-Gallo, et al., “Unveiling the Epigenetic Impact of Vegan vs. omnivorous Diets on Aging: Insights From the Twins Nutrition Study (TwiNS),” BMC Medicine [Electronic Resource] 22 (2024): 301.

[371]

H. Bischoff-Ferrari, S. Gangler, M. Wieczorek, et al., “Individual and Additive Effects of Vitamin D, Omega-3 and Exercise on DNA Methylation Clocks of Biological Aging in Older Adults From the DO-HEALTH Trial,” Nat Aging 5 (2025): 376-385.

[372]

J. Chen, J. Xiang, M. Zhou, et al., “Dietary Timing Enhances Exercise by Modulating Fat-muscle Crosstalk via Adipocyte AMPKalpha2 Signaling,” Cell metabolism (2025), https://doi.org/10.1016/j.cmet.2025.02.007.

[373]

E. Sutton, R. Beyl, K. Early, W. Cefalu, E. Ravussin, and C. Peterson, “Early Time-Restricted Feeding Improves Insulin Sensitivity, Blood Pressure, and Oxidative Stress Even Without Weight Loss in Men With Prediabetes,” Cell metabolism 27 (2018): 1212-1221. e1213.

[374]

J. Cavalcanti de Albuquerque, J. Hunter, R. Domingues, et al., “Brain Sensing of Metabolic state Regulates Circulating Monocytes,” Science Immunology 10 (2025): eadr3226.

[375]

F. Fox, D. Liu, M. Breteler, and N. Aziz, “Physical Activity Is Associated With Slower Epigenetic Ageing-Findings From the Rhineland Study,” Aging Cell 22 (2023): e13828.

[376]

A. Keiser, T. Dong, E. Kramar, et al., “Specific Exercise Patterns Generate an Epigenetic Molecular Memory Window That Drives Long-term Memory Formation and Identifies ACVR1C as a Bidirectional Regulator of Memory in Mice,” Nature Communications 15 (2024): 3836.

[377]

H. Lee, B. Kim, and T. Park, “The Association Between Sleep Quality and Accelerated Epigenetic Aging With Metabolic Syndrome in Korean Adults,” Clin Epigenetics 16 (2024): 92.

[378]

K. Fitzgerald, R. Hodges, D. Hanes, et al., “Potential Reversal of Epigenetic Age Using a Diet and Lifestyle Intervention: A Pilot Randomized Clinical Trial,” Aging (Albany NY) 13 (2021): 9419-9432.

[379]

H. Xu, O. Li, D. Kim, Z. Bao, and F. Yang, “Gut Microbiota and Epigenetic Age Acceleration: A bi-directional Mendelian Randomization Study,” Aging Clinical and Experimental Research 36 (2024): 227.

[380]

K. Kim, Y. Chung, J. Huh, et al., “Gut Microbiota of the Young Ameliorates Physical Fitness of the Aged in Mice,” Microbiome 10 (2022): 238.

[381]

T. Ghosh, F. Shanahan, and P. O'Toole, “The Gut Microbiome as a Modulator of Healthy Ageing,” Nature reviews Gastroenterology & hepatology 19 (2022): 565-584.

[382]

T. Liu, D. Du, R. Zhao, Q. Xie, and Z. Dong, “Gut Microbes Influence the Development of central Nervous System Disorders Through Epigenetic Inheritance,” Microbiological Research 274 (2023): 127440.

[383]

Y. Jing, X. Jiang, Q. Ji, et al., “Genome-wide CRISPR Activation Screening in Senescent Cells Reveals SOX5 as a Driver and Therapeutic Target of Rejuvenation,” Cell Stem Cell 30 (2023): 1452-1471. e1410.

[384]

B. Zhang, D. Lee, A. Trapp, et al., “Multi-omic Rejuvenation and Life Span Extension on Exposure to Youthful Circulation,” Nat Aging 3 (2023): 948-964.

[385]

J. Poganik, B. Zhang, G. Baht, et al., “Biological Age Is Increased by Stress and Restored Upon Recovery,” Cell metabolism 35 (2023): 807-820. e805.

[386]

J. Sanz-Ros, N. Romero-Garcia, C. Mas-Bargues, et al., “Small Extracellular Vesicles From Young Adipose-derived Stem Cells Prevent Frailty, Improve Health Span, and Decrease Epigenetic Age in Old Mice,” Science Advances 8 (2022): eabq2226.

[387]

F. Galkin, P. Mamoshina, K. Kochetov, D. Sidorenko, and A. Zhavoronkov, “DeepMAge: A Methylation Aging Clock Developed With Deep Learning,” Aging Dis 12 (2021): 1252-1262.

[388]

A. Teschendorff and S. Horvath, “Epigenetic Ageing Clocks: Statistical Methods and Emerging Computational Challenges,” Nature Reviews Genetics 26 (2025): 350-368.

[389]

F. Prattichizzo, C. Frige, V. Pellegrini, et al., “Organ-specific Biological Clocks: Ageotyping for Personalized Anti-aging Medicine,” Ageing Research Reviews 96 (2024): 102253.

[390]

M. Buckley, E. Sun, B. George, et al., “Cell-type-specific Aging Clocks to Quantify Aging and Rejuvenation in Neurogenic Regions of the Brain,” Nat Aging 3 (2023): 121-137.

[391]

K. Browder, P. Reddy, M. Yamamoto, et al., “In Vivo Partial Reprogramming Alters Age-associated Molecular Changes During Physiological Aging in Mice,” Nat Aging 2 (2022): 243-253.

[392]

A. Yucel and V. Gladyshev, “The Long and Winding Road of Reprogramming-induced Rejuvenation,” Nature Communications 15 (2024): 1941.

[393]

D. Gill, A. Parry, F. Santos, et al., “Multi-omic Rejuvenation of human Cells by Maturation Phase Transient Reprogramming,” Elife 11 (2022): e71624.

[394]

G. Lopez-Lluch, N. Hunt, B. Jones, et al., “Calorie Restriction Induces Mitochondrial Biogenesis and Bioenergetic Efficiency,” PNAS 103 (2006): 1768-1773.

[395]

J. Yoshino, J. Baur, and S. Imai, “NAD(+) Intermediates: The Biology and Therapeutic Potential of NMN and NR,” Cell metabolism 27 (2018): 513-528.

[396]

S. Smallwood, H. Lee, C. Angermueller, et al., “Single-cell Genome-wide Bisulfite Sequencing for Assessing Epigenetic Heterogeneity,” Nature Methods 11 (2014): 817-820.

[397]

O. Schwartzman and A. Tanay, “Single-cell Epigenomics: Techniques and Emerging Applications,” Nature Reviews Genetics 16 (2015): 716-726.

[398]

D. Rossi, D. Bryder, J. Zahn, et al., “Cell Intrinsic Alterations Underlie Hematopoietic Stem Cell Aging,” PNAS 102 (2005): 9194-9199.

[399]

X. Li, X. Zeng, Y. Xu, et al., “Mechanisms and Rejuvenation Strategies for Aged Hematopoietic Stem Cells,” Journal of hematology & oncology 13 (2020): 31.

[400]

N. Habib, I. Avraham-Davidi, A. Basu, et al., “Massively Parallel Single-nucleus RNA-seq With DroNc-seq,” Nature Methods 14 (2017): 955-958.

[401]

Z. Shi, Y. Geng, J. Liu, et al., “Single-cell Transcriptomics Reveals Gene Signatures and Alterations Associated With Aging in Distinct Neural Stem/Progenitor Cell Subpopulations,” Protein Cell 9 (2018): 351-364.

[402]

D. Puri and W. Wagner, “Epigenetic Rejuvenation by Partial Reprogramming,” BioEssays 45 (2023): e2200208.

[403]

B. Grigorash, D. van Essen, G. Liang, et al., “p16(High) senescence Restricts Cellular Plasticity During Somatic Cell Reprogramming,” Nature Cell Biology 25 (2023): 1265-1278.

[404]

B. Wang, J. Han, J. Elisseeff, and M. Demaria, “The Senescence-associated Secretory Phenotype and Its Physiological and Pathological Implications,” Nature Reviews Molecular Cell Biology 25 (2024): 958-978.

[405]

M. Baar, R. Brandt, D. Putavet, et al., “Targeted Apoptosis of Senescent Cells Restores Tissue Homeostasis in Response to Chemotoxicity and Aging,” Cell 169 (2017): 132-147. e116.

[406]

Y. Lai, I. Ramirez-Pardo, J. Isern, et al., “Multimodal Cell Atlas of the Ageing human Skeletal Muscle,” Nature 629 (2024): 154-164.

[407]

V. Moiseeva, A. Cisneros, V. Sica, et al., “Senescence Atlas Reveals an Aged-Like Inflamed Niche That Blunts Muscle Regeneration,” Nature 613 (2023): 169-178.

[408]

S. Bocklandt, W. Lin, M. Sehl, et al., “Epigenetic Predictor of Age,” PLoS ONE 6 (2011): e14821.

[409]

M. Levine, “Modeling the Rate of Senescence: Can Estimated Biological Age Predict Mortality More Accurately Than Chronological Age?” Journals of Gerontology. Series A, Biological Sciences and Medical Sciences 68 (2013): 667-674.

[410]

B. Joyce, T. Gao, Y. Zheng, et al., “Epigenetic Age Acceleration Reflects Long-Term Cardiovascular Health,” Circulation Research 129 (2021): 770-781.

[411]

D. Belsky, A. Caspi, R. Houts, et al., “Quantification of Biological Aging in Young Adults,” PNAS 112 (2015): E4104-E4110.

[412]

E. Sillanpaa, A. Heikkinen, A. Kankaanpaa, et al., “Blood and Skeletal Muscle Ageing Determined by Epigenetic Clocks and Their Associations With Physical Activity and Functioning,” Clin Epigenetics 13 (2021): 110.

[413]

T. Tanaka, N. Basisty, G. Fantoni, et al., “Plasma Proteomic Biomarker Signature of Age Predicts Health and Life Span,” Elife 9 (2020): e61073.

[414]

R. Jansen, L. Han, J. Verhoeven, et al., “An Integrative Study of Five Biological Clocks in Somatic and Mental Health,” Elife 10 (2021): e59479.

[415]

W. Dai, X. Qiao, Y. Fang, et al., “Epigenetics-targeted Drugs: Current Paradigms and Future Challenges,” Signal Transduct Target Ther 9 (2024): 332.

[416]

S. McCutcheon, D. Rohm, N. Iglesias, and C. Gersbach, “Epigenome Editing Technologies for Discovery and Medicine,” Nature Biotechnology 42 (2024): 1199-1217.

[417]

X. Yu, H. Zhao, R. Wang, et al., “Cancer Epigenetics: From Laboratory Studies and Clinical Trials to Precision Medicine,” Cell Death Discov 10 (2024): 28.

[418]

X. Xu and L. Qi, “A CRISPR-dCas Toolbox for Genetic Engineering and Synthetic Biology,” Journal of Molecular Biology 431 (2019): 34-47.

[419]

P. Jogam, D. Sandhya, A. Alok, V. Peddaboina, V. Allini, and B. Zhang, “A Review on CRISPR/Cas-based Epigenetic Regulation in Plants,” International Journal of Biological Macromolecules 219 (2022): 1261-1271.

[420]

S. Xu, J. Kim, Q. Tang, et al., “CAS9 is a Genome Mutator by Directly Disrupting DNA-PK Dependent DNA Repair Pathway,” Protein Cell 11 (2020): 352-365.

[421]

F. Adikusuma, S. Piltz, M. Corbett, et al., “Large Deletions Induced by Cas9 Cleavage,” Nature 560 (2018): E8-E9.

[422]

S. Ling, X. Zhang, Y. Dai, et al., “Customizable Virus-Like Particles Deliver CRISPR-Cas9 ribonucleoprotein for Effective Ocular Neovascular and Huntington's disease Gene Therapy,” Nature Nanotechnology 20 (2025): 543-553.

[423]

X. Bian, L. Zhou, Z. Luo, et al., “Emerging Delivery Systems for Enabling Precision Nucleic Acid Therapeutics,” ACS Nano 19 (2025): 4039-4083.

[424]

Y. Zuo, C. Zhang, Y. Zhou, et al., “Liver-specific in Vivo Base Editing of Angptl3 via AAV Delivery Efficiently Lowers Blood Lipid Levels in Mice,” Cell Biosci 13 (2023): 109.

[425]

C. Aging Biomarker, H. Bao, J. Cao, et al., “Biomarkers of Aging,” Sci China Life Sci 66 (2023): 893-1066.

[426]

A. Kane and D. Sinclair, “Epigenetic Changes During Aging and Their Reprogramming Potential,” Critical Reviews in Biochemistry and Molecular Biology 54 (2019): 61-83.

[427]

S. Liesenfelder, M. Elsafi Mabrouk, J. Iliescu, et al., “Epigenetic Editing at Individual Age-associated CpGs Affects the Genome-wide Epigenetic Aging Landscape,” Nat Aging 5, no. 6 (2025): 997-1009, https://doi.org/10.1038/s43587-025-00841-1.

[428]

M. Ehrlich, “DNA Hypermethylation in Disease: Mechanisms and Clinical Relevance,” Epigenetics 14 (2019): 1141-1163.

[429]

S. Yi and K. Kim, “New Insights Into the Role of Histone Changes in Aging,” International Journal of Molecular Sciences 21 (2020): 8241.

[430]

F. Rossiello, D. Jurk, J. Passos, and F. d'Adda di Fagagna, “Telomere Dysfunction in Ageing and Age-related Diseases,” Nature Cell Biology 24 (2022): 135-147.

[431]

J. Amorim, G. Coppotelli, A. Rolo, C. Palmeira, J. Ross, and D. Sinclair, “Mitochondrial and Metabolic Dysfunction in Ageing and Age-related Diseases,” Nature reviews Endocrinology 18 (2022): 243-258.

[432]

D. Cheishvili, L. Boureau, and M. Szyf, “DNA Demethylation and Invasive Cancer: Implications for Therapeutics,” British Journal of Pharmacology 172 (2015): 2705-2715.

[433]

A. Ocampo, P. Reddy, and J. Belmonte, “Anti-Aging Strategies Based on Cellular Reprogramming,” Trends in Molecular Medicine 22 (2016): 725-738.

[434]

S. Velychko, K. Adachi, K. Kim, et al., “Excluding Oct4 From Yamanaka Cocktail Unleashes the Developmental Potential of iPSCs,” Cell Stem Cell 25 (2019): 737-753. e734.

[435]

J. Wu, T. Shao, Z. Tang, et al., “Highly Efficient Construction of Monkey Blastoid Capsules From Aged Somatic Cells,” Nature Communications 16 (2025): 1130.

[436]

W. Dong, S. Liu, S. Li, and Z. Wang, “Cell Reprogramming Therapy for Parkinson's Disease,” Neural Regen Res 19 (2024): 2444-2455.

[437]

S. Wen, R. Zheng, C. Cai, and W. Jiang, “Chemical-based Epigenetic Reprogramming to Advance Pluripotency and Totipotency,” Nature Chemical Biology 21, no. 5 (2025): 635-647, https://doi.org/10.1038/s41589-025-01874-8.

[438]

M. Habets, M. Rots, and L. Chiapperino, “Meeting Report on the Round Table Discussions ‘Epigenetics and Society’ CLEPIC24,” Epigenetics Communications 5 (2025), https://doi.org/10.1186/s43682-025-00035-1.

[439]

T. Tollefsbol, “Advances in Epigenetic Technology,” Methods in Molecular Biology 791 (2011): 1-10.

[440]

N. Rivero-Segura, O. Bello-Chavolla, O. Barrera-Vazquez, L. Gutierrez-Robledo, and J. Gomez-Verjan, “Promising Biomarkers of human Aging: In Search of a Multi-omics Panel to Understand the Aging Process From a Multidimensional Perspective,” Ageing Research Reviews 64 (2020): 101164.

[441]

F. Xiao, H. Wang, L. Zhao, et al., “Methylome Analysis in Long-lived Men Deciphers DNA Methylation Modifications Associated With Male Longevity in Humans,” Cell reports 44 (2025): 115158.

[442]

W. Xie, Y. Ke, Q. You, et al., “Single-Cell RNA Sequencing and Assay for Transposase-Accessible Chromatin Using Sequencing Reveals Cellular and Molecular Dynamics of Aortic Aging in Mice,” Arteriosclerosis, Thrombosis, and Vascular Biology 42 (2022): 156-171.

[443]

K. Miskimen, E. Chan, and J. Haines, “Assay for Transposase-Accessible Chromatin Using Sequencing (ATAC-seq) Data Analysis,” Current protocols in human genetics 92 (2017), https://doi.org/10.1002/cphg.32. 20 24 21-20 24 13.

[444]

I. Beerman and D. Rossi, “Epigenetic Control of Stem Cell Potential During Homeostasis, Aging, and Disease,” Cell Stem Cell 16 (2015): 613-625.

[445]

Y. Gao, Y. Chi, Y. Chen, et al., “Multi-omics Analysis of human Mesenchymal Stem Cells Shows Cell Aging That Alters Immunomodulatory Activity Through the Downregulation of PD-L1,” Nature Communications 14 (2023): 4373.

[446]

J. Si, Y. Ma, C. Yu, et al., “DNA Methylation Age Mediates Effect of Metabolic Profile on Cardiovascular and General Aging,” Circulation Research 135 (2024): 954-966.

[447]

Z. Zheng, J. Li, T. Liu, et al., “DNA Methylation Clocks for Estimating Biological Age in Chinese Cohorts,” Protein Cell 15 (2024): 575-593.

[448]

J. Rutledge, H. Oh, and T. Wyss-Coray, “Measuring Biological Age Using Omics Data,” Nature Reviews Genetics 23 (2022): 715-727.

[449]

H. Huang, Y. Chen, W. Xu, et al., “Decoding Aging Clocks: New Insights From Metabolomics,” Cell metabolism 37 (2025): 34-58.

[450]

K. Ying, H. Liu, A. Tarkhov, et al., “Causality-enriched Epigenetic Age Uncouples Damage and Adaptation,” Nat Aging 4 (2024): 231-246.

[451]

M. He, X. Zhou, Z. Li, et al., “Programmable Transcriptional Modulation With a Structured RNA-Mediated CRISPR-dCas9 Complex,” Journal of the American Chemical Society 144 (2022): 12690-12697.

[452]

M. Cappelluti, V. Mollica Poeta, S. Valsoni, et al., “Durable and Efficient Gene Silencing in Vivo by Hit-and-run Epigenome Editing,” Nature 627 (2024): 416-423.

[453]

Y. Wang, R. Huang, S. Feng, and R. Mo, “Advances in Nanocarriers for Targeted Drug Delivery and Controlled Drug Release,” Chin J Nat Med 23 (2025): 513-528.

[454]

C. Policarpi, M. Munafo, S. Tsagkris, V. Carlini, and J. Hackett, “Systematic Epigenome Editing Captures the Context-dependent Instructive Function of Chromatin Modifications,” Nature Genetics 56 (2024): 1168-1180.

[455]

A. Majchrzak-Celinska, A. Warych, and M. Szoszkiewicz, “Novel Approaches to Epigenetic Therapies: From Drug Combinations to Epigenetic Editing,” Genes (Basel) 12 (2021): 208.

[456]

D. Burdusel, T. Doeppner, R. Surugiu, D. Hermann, D. Olaru, and A. Popa-Wagner, “The Intersection of Epigenetics and Senolytics in Mechanisms of Aging and Therapeutic Approaches,” Biomolecules 15 (2024): 18.

[457]

J. de Toro-Martin, B. Arsenault, J. Despres, and M. Vohl, “Precision Nutrition: A Review of Personalized Nutritional Approaches for the Prevention and Management of Metabolic Syndrome,” Nutrients 9 (2017): 913.

[458]

R. Nativio, Y. Lan, G. Donahue, et al., “An Integrated Multi-omics Approach Identifies Epigenetic Alterations Associated With Alzheimer's Disease,” Nature Genetics 52 (2020): 1024-1035.

[459]

A. Herman, J. Occean, and P. Sen, “Epigenetic Dysregulation in Cardiovascular Aging and Disease,” J Cardiovasc Aging 1 (2021),

RIGHTS & PERMISSIONS

2025 The Author(s). MedComm published by Sichuan International Medical Exchange & Promotion Association (SCIMEA) and John Wiley & Sons Australia, Ltd.

AI Summary AI Mindmap
PDF

14

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/